PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 3140993-14 1988 Our study supports the theory of more than one type of UDP-GT being involved in morphine glucuronidation. Morphine 80-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 33865918-5 2021 In response to three model pro-genotoxicants: benzo[a]pyrene, Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and 2-aminoanthracene (2-AA),we observed further transcriptional upregulation of xenobiotic metabolising genes (CYP1A1, CYP1A2, NAT1/2, SULT1A2, UGT1A1, UGT1A3) compared to untreated spheroids. amino-1-methyl-6-phenylimidazo[4,5-b]pyridine 62-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 260-266 807500-1 1975 Reduction in caloric intake was associated with a greater absolute rise in the serum bilirubin concentration in patients with Gilbert"s syndrome and partial hepatic bilirubin uridine diphosphate glucuronyltransferase (UDPG-T) dysfunction compared to patients with hemolytic unconjugated hyperbilirubinemia and normal subjects. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 175-216 33785516-13 2021 Significance Statement The metabolism and excretion of darolutamide in humans revealed that oxidation (CYP3A4) and glucuronidation (UGT1A9, UGT1A1) were the main metabolic routes of elimination. darolutamide 55-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 34044055-0 2021 In vitro inhibition of human UDP-glucuronosyltransferase (UGT) 1A1 by osimertinib, and prediction of in vivo drug-drug interactions. osimertinib 70-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-66 34044055-3 2021 High inhibitory effect of osimertinib was shown towards UGT1A1, 1A3, 1A6, 1A7, 1A8, 1A10, 2B7 and 2B15. osimertinib 26-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 34044055-4 2021 Especially, osimertinib exhibited competitive inhibition against UGT1A1 with a Ki,u of 0.87 +- 0.12 muM. osimertinib 12-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 34044055-6 2021 Results from quantitative prediction study indicated that osimertinib administered at 80 mg/day may result in a 4.83% increase in the AUC of drugs mainly metabolized by UGT1A1, implying low risk of DDI via liver metabolism. osimertinib 58-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 34038714-3 2021 Our data indicated that ibrutinib exerted broad inhibition on most of UGTs, including a potent competitive inhibition against UGT1A1 with a Ki value of 0.90 +- 0.03 muM, a noncompetitive inhibition against UGT1A3 and UGT1A7 with Ki values of 0.88 +- 0.03 muM and 2.52 +- 0.23 muM, respectively, while acalabrutinib only exhibited weak UGT inhibition towards all tested UGT isoforms. ibrutinib 24-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 33877904-0 2021 The Correlation Between UGT1A1 Gene Phenotypes and the Clinical Prognosis of Advanced Colorectal Cancer After FOLFIRI Therapy. FOLFIRI regimen 110-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 34038714-4 2021 DDI risk prediction suggested that the inhibition against UGT1A1 and UGT1A3 by ibrutinib might bring a potential DDIs risk, while acalabrutinib was unlikely to trigger clinically significant UGT-mediated DDIs due to its weak effects. ibrutinib 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 33610566-8 2021 Moreover, isobavachalcone demonstrated broad-spectrum inhibitory effects against CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, UGT1A1, UGT1A9, UGT2B7 with IC50 values of 1.08-9.78 muM. isobavachalcone 10-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 34007799-0 2021 UGT1A1-related Bilirubin Encephalopathy/Kernicterus in Adults. Bilirubin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33901188-10 2021 We replicated several proof-of-concept transcriptionally regulated gene-trait associations, including UGT1A1 (encoding bilirubin uridine diphosphate glucuronosyltransferase enzyme) and total bilirubin levels (p = 3.59x10-12), and CETP (cholesteryl ester transfer protein) with high-density lipoprotein cholesterol (p = 4.49x10-12). Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 33901188-10 2021 We replicated several proof-of-concept transcriptionally regulated gene-trait associations, including UGT1A1 (encoding bilirubin uridine diphosphate glucuronosyltransferase enzyme) and total bilirubin levels (p = 3.59x10-12), and CETP (cholesteryl ester transfer protein) with high-density lipoprotein cholesterol (p = 4.49x10-12). Uridine Diphosphate 129-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 33485974-4 2021 METHODS: RT-qPCR was used to detect the effects of water extract of Polygoni Multiflori Radix (PMR) and its main components on the mRNA expression of CYP3A4 and UGT1A1 in human hepatic parenchyma cell line L02. Water 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 33485974-18 2021 CONCLUSION: The inhibition of mRNA expression of CYP3A4 or UGT1A1 enhanced hepatotoxicity of PMR. pmr 93-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 33631237-3 2021 The present study aimed to determine the genotype of the UGT1A1 promoter and exon that are related to the serum total bilirubin (STB) level in the Chinese Han population. Bilirubin 118-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 33995076-9 2021 Our findings demonstrate that PRH alter expression and function of UGT proteins in an isoform-specific manner and increase UGT1A1-mediated labetalol metabolism in human hepatocytes by inducing UGT1A1 protein concentrations. Labetalol 139-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 33995076-9 2021 Our findings demonstrate that PRH alter expression and function of UGT proteins in an isoform-specific manner and increase UGT1A1-mediated labetalol metabolism in human hepatocytes by inducing UGT1A1 protein concentrations. Labetalol 139-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 33995076-0 2021 Pregnancy-Related Hormones Increase UGT1A1-Mediated Labetalol Metabolism in Human Hepatocytes. Labetalol 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 33995076-3 2021 Using primary human hepatocytes, we evaluated the effect of PRH exposure on mRNA levels and protein concentrations of UGT1A1, UGT2B7, and other key UGT enzymes, and on the metabolism of labetalol (a UGT1A1 and UGT2B7 substrate commonly prescribed to treat hypertensive disorders of pregnancy). Labetalol 186-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 33830459-6 2021 Cabotegravir is mainly metabolized by UGT1A1, and rilpivirine is mainly metabolized by CYP3A4, therefore these agents are susceptible to DDIs with inhibitors, and particularly inducers of drug-metabolizing enzymes. cabotegravir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 33995076-7 2021 PRH exposure also significantly increased labetalol metabolism to its UGT1A1-derived glucuronide metabolite in a concentration-dependent manner, which positively correlated with PRH-induced changes in UGT1A1 protein concentrations. Labetalol 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 33995076-7 2021 PRH exposure also significantly increased labetalol metabolism to its UGT1A1-derived glucuronide metabolite in a concentration-dependent manner, which positively correlated with PRH-induced changes in UGT1A1 protein concentrations. Labetalol 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 33995076-7 2021 PRH exposure also significantly increased labetalol metabolism to its UGT1A1-derived glucuronide metabolite in a concentration-dependent manner, which positively correlated with PRH-induced changes in UGT1A1 protein concentrations. Glucuronides 85-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 33995076-7 2021 PRH exposure also significantly increased labetalol metabolism to its UGT1A1-derived glucuronide metabolite in a concentration-dependent manner, which positively correlated with PRH-induced changes in UGT1A1 protein concentrations. Glucuronides 85-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 33837924-7 2021 Reports for UGT1A1-metabolized irinotecan and the CYP2C19-metabolized drugs voriconazole, escitalopram and clopidogrel were selected for comparison between the subgroups based upon known genetic polymorphisms with high prevalence in Japan. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 33733372-3 2021 METHODS: Two pharmacokinetic and one myelosuppression model were assembled to predict concentrations of irinotecan and its metabolite SN-38 given different UGT1A1 genotypes (poor metabolizers: CLSN-38: -36%) and neutropenia following conventional versus PGx-based dosing (350 versus 245 mg/m2 (-30%)). Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 33759451-7 2021 Subsequently, the model was used to explore UGT1A1-related DDI risk with atazanavir and rifampicin along with the effect of enzyme genotype on raltegravir apparent clearance. Atazanavir Sulfate 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 33759451-7 2021 Subsequently, the model was used to explore UGT1A1-related DDI risk with atazanavir and rifampicin along with the effect of enzyme genotype on raltegravir apparent clearance. Rifampin 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Rifampin 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 33759451-8 2021 Simulations of pregnancy-related induction of UGT1A1 either exacerbated UGT1A1 induction by rifampicin or negated atazanavir UGT1A1 inhibition. Atazanavir Sulfate 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 33462346-2 2021 DPYD and UGT1A1 variants are relevant predictors of fluoropyrimidine and irinotecan-associated adverse events (AEs). 2-fluoropyrimidine 52-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 33462346-2 2021 DPYD and UGT1A1 variants are relevant predictors of fluoropyrimidine and irinotecan-associated adverse events (AEs). Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 33462346-9 2021 DPYD IVS14+1A allele is confirmed to be associated with fluoropyrimidine life-threatening toxicities, and UGT1A1*28 is related with a higher risk of hematologic AEs following irinotecan treatment. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 33024987-6 2021 Increases in total bilirubin were more frequently observed in patients treated with enasidenib, consistent with this inhibitor"s known potential to inhibit UGT1A1, but did not appear to have significant clinical consequences. enasidenib 84-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 33733372-3 2021 METHODS: Two pharmacokinetic and one myelosuppression model were assembled to predict concentrations of irinotecan and its metabolite SN-38 given different UGT1A1 genotypes (poor metabolizers: CLSN-38: -36%) and neutropenia following conventional versus PGx-based dosing (350 versus 245 mg/m2 (-30%)). Irinotecan 134-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 33290829-3 2021 Belinostat exhibited a wide range of inhibition against UGTs activities, particularly a potent non-competitive inhibition against UGT1A3, and weak inhibition against UGT1A1, 1A7, 1A8, 2B4 and 2B7. belinostat 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 33790625-0 2021 Influence of UGT1A1 *6/*28 Polymorphisms on Irinotecan-Related Toxicity and Survival in Pediatric Patients with Relapsed/Refractory Solid Tumors Treated with the VIT Regimen. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 33790625-1 2021 Objective: The association between UGT1A1*6/*28 polymorphisms and treatment outcomes of irinotecan in children remains unknown. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 33790625-2 2021 This retrospective study investigated the influence of UGT1A1*6/*28 polymorphisms on irinotecan toxicity and survival of pediatric patients with relapsed/refractory solid tumors. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 33790625-8 2021 Moreover, patients with mutant UGT1A1*6 genotypes were more likely to develop grade I-IV irinotecan-related diarrhea (P = 0.043) and anemia (P = 0.002). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 33790625-9 2021 Overall, the UGT1A1*28 polymorphism may play a protective role against irinotecan-related diarrhea and abdominal pain. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 33790625-10 2021 Conclusion: In relapsed/refractory pediatric solid tumors, the UGT1A1*6 polymorphism was a risk factor of irinotecan-related diarrhea and anemia. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33790625-11 2021 The UGT1A1*28 polymorphism may serve a protective role in irinotecan-related abdominal pain and diarrhea. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 33386925-0 2021 FOLFIRINOX in advanced pancreatic cancer patients with the double-variant type of UGT1A1 *28 and *6 polymorphism: a multicenter, retrospective study. folfirinox 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-1 2021 BACKGROUND: UGT1A1 *28 and *6 polymorphism is associated with reduced enzyme activity and severe toxicities of irinotecan, especially in patients with homozygous or heterozygous for UGT1A1*28 or *6 polymorphism for both UGT1A1*28 and *6 (double-variant-type of UGT1A1 polymorphism, UGT1A1-DV). Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 33386925-3 2021 The optimal dose of irinotecan as a component of the FOLFIRINOX has not been established yet for patients with UGT1A1-DV. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 33386925-5 2021 We evaluated the patient characteristics, efficacy and safety of FOLFIRINOX and investigate the optimal initial dose of irinotecan in Japanese advanced PC patients with UGT1A1-DV. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 33619631-0 2021 Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 33619631-0 2021 Effect of UGT1A1, CYP3A and CES Activities on the Pharmacokinetics of Irinotecan and its Metabolites in Patients with UGT1A1 Gene Polymorphisms. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 33619631-2 2021 SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 33619631-2 2021 SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). 7-ethyl-10-hydroxycamptothecin glucuronide 51-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 33619631-2 2021 SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). 7-ethyl-10-hydroxycamptothecin glucuronide 70-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 33619631-3 2021 Genetic polymorphisms in UGT1A1 have been associated with altered SN-38 pharmacokinetics, which increase the risk of toxicity in patients. Irinotecan 66-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 33619631-5 2021 We evaluated the activities of UGT1A1, CYP3A, and CES and the factors affecting the pharmacokinetics of plasma SN-38 in patients with UGT1A1 gene polymorphisms. Irinotecan 111-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 33619631-12 2021 Since UGT1A1 activity was low in patient 1 with a high AUC of SN-38, it can be said that the increase in plasma concentration was due to a decrease in UGT1A1 activity. Irinotecan 62-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. 7-ethyl-10-hydroxycamptothecin glucuronide 86-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 33619631-15 2021 CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Irinotecan 86-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 33728696-0 2021 UGT1A1 Gene Polymorphisms in Patients with Small Cell Lung Cancer Treated with Irinotecan-Platinum Doublet Chemotherapy and Their Association with Gastrointestinal Toxicity and Overall Survival. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33728696-0 2021 UGT1A1 Gene Polymorphisms in Patients with Small Cell Lung Cancer Treated with Irinotecan-Platinum Doublet Chemotherapy and Their Association with Gastrointestinal Toxicity and Overall Survival. Platinum 90-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33728696-18 2021 IMPLICATIONS FOR PRACTICE: UGT1A1 gene polymorphisms are known to influence irinotecan-related toxicity. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Platinum 105-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Platinum 105-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33728696-19 2021 In this prospective cohort study of patients with small cell lung cancer receiving first-line irinotecan-platinum chemotherapy, the prevalence of UGT1A1*6 polymorphisms was found to be 10.8% UGT1A1*6 and 58.8% UGT1A1*28 homo/heterozygous mutant, respectively. Platinum 105-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 33386925-9 2021 CONCLUSION: Based on our findings, we recommend that in Japanese advanced PC patients with UGT1A1- DV treated with FOLFIRINOX, irinotecan be administered at an initial dose of <= 120 mg/m2. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 33376146-0 2021 Detoxication vs. Bioactivation Pathways of Lapatinib In Vitro: UGT1A1 Catalyzes the Hepatic Glucuronidation of Debenzylated Lapatinib. Lapatinib 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33376146-0 2021 Detoxication vs. Bioactivation Pathways of Lapatinib In Vitro: UGT1A1 Catalyzes the Hepatic Glucuronidation of Debenzylated Lapatinib. Lapatinib 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33376146-4 2021 In the present study, reaction phenotyping experiments using human recombinant UGT enzymes and enzyme-selective chemical inhibitors demonstrated that UGT1A1 was the major hepatic UGT enzyme involved in lapatinib M1 glucuronidation. Lapatinib 202-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 33376146-5 2021 Formation of the M1-glucuronide by human liver microsomes from UGT1A1-genotyped donors was significantly correlated with UGT1A1 activity, as measured by 17beta-estradiol 3-glucuronidation (R2 = 0.90). m1-glucuronide 17-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33376146-5 2021 Formation of the M1-glucuronide by human liver microsomes from UGT1A1-genotyped donors was significantly correlated with UGT1A1 activity, as measured by 17beta-estradiol 3-glucuronidation (R2 = 0.90). m1-glucuronide 17-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 33376146-5 2021 Formation of the M1-glucuronide by human liver microsomes from UGT1A1-genotyped donors was significantly correlated with UGT1A1 activity, as measured by 17beta-estradiol 3-glucuronidation (R2 = 0.90). Estradiol 153-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33376146-5 2021 Formation of the M1-glucuronide by human liver microsomes from UGT1A1-genotyped donors was significantly correlated with UGT1A1 activity, as measured by 17beta-estradiol 3-glucuronidation (R2 = 0.90). Estradiol 153-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 33376146-12 2021 UGT1A1 was identified as the major hepatic enzyme involved in lapatinib M1 glucuronidation, which may limit hepatic exposure to the potentially toxic quinoneimine. Lapatinib 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33376146-12 2021 UGT1A1 was identified as the major hepatic enzyme involved in lapatinib M1 glucuronidation, which may limit hepatic exposure to the potentially toxic quinoneimine. quinoneimine 150-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33549631-6 2021 UGT1A1 and -1A10 were inhibited by TPT, whereas UGT 2B4 and -2B7 were inhibited by TBT. triphenyltin 35-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-16 33549631-10 2021 TPT can additionally inhibit activities of UGT1A1 and -1A10 in estradiol-3-O-glucuronidation, with IC50 values of a few micro-molars. triphenyltin 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-59 33549631-10 2021 TPT can additionally inhibit activities of UGT1A1 and -1A10 in estradiol-3-O-glucuronidation, with IC50 values of a few micro-molars. Estradiol 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-59 33290829-5 2021 Our data indicate that the intravenous infusion of belinostat at clinical available dose can contribute a significant increase to the AUC of co-administrated drugs primarily cleared by UGT1A3 or UGT1A1, which will result in potential DDIs. belinostat 51-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 195-201 33263770-3 2021 The effects of a series of di- and trihydroxychalcones as AhR agonists was structure-dependent with maximal induction of CYP1A1, CYP1B1 and UGT1A1 in Caco2 cells observed for compounds containing 2,2"-dihydroxy substituents and this included 2,2"-dihydroxy-, 2,2",4"-trihydroxy- and 2,2",5"-trihydroxychalcones. di- and trihydroxychalcones 27-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 33263770-3 2021 The effects of a series of di- and trihydroxychalcones as AhR agonists was structure-dependent with maximal induction of CYP1A1, CYP1B1 and UGT1A1 in Caco2 cells observed for compounds containing 2,2"-dihydroxy substituents and this included 2,2"-dihydroxy-, 2,2",4"-trihydroxy- and 2,2",5"-trihydroxychalcones. 2,2"-dihydroxy 196-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 33622223-6 2021 RESULTS: Letermovir was a substrate of CYP3A4/5 and UGT1A1/3 in vitro. letermovir 9-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-60 33626178-1 2021 Bictegravir is equally metabolized by cytochrome P-450 (CYP)3A and uridine diphosphate-glucuronosyltransferase (UGT)1A1. bictegravir 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-119 33622223-10 2021 Letermovir inhibited CYP2B6, CYP2C8, CYP3A, and UGT1A1 in vitro, and induced CYP3A4 and CYP2B6 in hepatocytes. letermovir 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 33685083-1 2021 Objective: To investigate the diagnosis method of Gilbert syndrome (GS) and the relationship between UGT1A1 gene polymorphism distribution with serum bilirubin. Bilirubin 150-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 33685083-6 2021 Among 110 cases with Gilbert syndrome combined with non-hemolytic diseases, pairwise comparisons showed that the total bilirubin levels of patients with UGT1A1*28/*28 mutations were significantly higher than UGT1A1*6/*6 and UGT1A1*1/*28 + *1/*6 mutation (P < 0.05). Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 33685083-6 2021 Among 110 cases with Gilbert syndrome combined with non-hemolytic diseases, pairwise comparisons showed that the total bilirubin levels of patients with UGT1A1*28/*28 mutations were significantly higher than UGT1A1*6/*6 and UGT1A1*1/*28 + *1/*6 mutation (P < 0.05). Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 208-214 33685083-6 2021 Among 110 cases with Gilbert syndrome combined with non-hemolytic diseases, pairwise comparisons showed that the total bilirubin levels of patients with UGT1A1*28/*28 mutations were significantly higher than UGT1A1*6/*6 and UGT1A1*1/*28 + *1/*6 mutation (P < 0.05). Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 208-214 33685083-7 2021 Additionally, with the increase of UGT1A1*28 distribution, the serum total bilirubin level had gradually increased (P = 0.028), but UGT1A1*6 was opposite (P = 0.021). Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 33628187-3 2020 This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Flavonoids 67-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 33628187-0 2020 Neobavaisoflavone Induces Bilirubin Metabolizing Enzyme UGT1A1 via PPARalpha and PPARgamma. neobavaisoflavone 0-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33551819-7 2020 Conversely, TCDD and OP, the activators of AhR, induced CYP1A1 (38 and 37-fold), and UGT1A1 (4.3 and 5.0-fold), respectively. Polychlorinated Dibenzodioxins 12-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 33628187-0 2020 Neobavaisoflavone Induces Bilirubin Metabolizing Enzyme UGT1A1 via PPARalpha and PPARgamma. Bilirubin 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33628187-1 2020 UDP-glucuronosyltransferase 1A1 (UGT1A1) is an essential enzyme in mammals that is responsible for detoxification and metabolic clearance of the endogenous toxin bilirubin and a variety of xenobiotics, including some crucial therapeutic drugs. Bilirubin 162-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 33628187-1 2020 UDP-glucuronosyltransferase 1A1 (UGT1A1) is an essential enzyme in mammals that is responsible for detoxification and metabolic clearance of the endogenous toxin bilirubin and a variety of xenobiotics, including some crucial therapeutic drugs. Bilirubin 162-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 33628187-3 2020 This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Flavonoids 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 33628187-3 2020 This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Flavonoids 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 33628187-3 2020 This study aims to find efficacious UGT1A1 inducer(s) from natural flavonoids, and to reveal the mechanism involved in up-regulating of this key conjugative enzyme by the flavonoid(s) with strong UGT1A1 induction activity. Flavonoids 67-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 33628187-4 2020 Among all the tested flavonoids, neobavaisoflavone (NBIF) displayed the most potent UGT1A1 induction activity, while its inductive effects were confirmed by both western blot and glucuronidation activity assays. neobavaisoflavone 33-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 33628187-4 2020 Among all the tested flavonoids, neobavaisoflavone (NBIF) displayed the most potent UGT1A1 induction activity, while its inductive effects were confirmed by both western blot and glucuronidation activity assays. neobavaisoflavone 52-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 33097971-2 2021 Our aim was to assess the efficacy and safety of induction treatment with FOLFOXIRI plus bevacizumab in patients with untreated mCRC harboring UGT1A1 wild (*1/*1), or single-hetero (*1/*6 or *1/*28) genotypes. folfoxiri 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 33278020-10 2021 Olanzapine and the UDP glucuronosyltransferase family 1 member A1 (UGT1A1) rs887829 polymorphism were associated with elevated glucose levels. Glucose 127-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-65 33278020-10 2021 Olanzapine and the UDP glucuronosyltransferase family 1 member A1 (UGT1A1) rs887829 polymorphism were associated with elevated glucose levels. Glucose 127-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 33535454-3 2021 Eudesmin, fargesin, epimagnolin A, magnolin, and yangambin inhibited UGT1A1 and UGT1A3 activities, but showed negligible inhibition of UGT1A4, UGT16, UGT1A9, and UGT2B7 activities at 200 muM in pooled human liver microsomes. magnolin 20-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 33535454-3 2021 Eudesmin, fargesin, epimagnolin A, magnolin, and yangambin inhibited UGT1A1 and UGT1A3 activities, but showed negligible inhibition of UGT1A4, UGT16, UGT1A9, and UGT2B7 activities at 200 muM in pooled human liver microsomes. magnolin 23-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 33535454-3 2021 Eudesmin, fargesin, epimagnolin A, magnolin, and yangambin inhibited UGT1A1 and UGT1A3 activities, but showed negligible inhibition of UGT1A4, UGT16, UGT1A9, and UGT2B7 activities at 200 muM in pooled human liver microsomes. epiyangambin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 33535454-4 2021 Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 muM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. magnolin 30-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 33535454-4 2021 Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 muM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. magnolin 30-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 232-238 33535454-4 2021 Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 muM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. magnolin 33-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 33535454-4 2021 Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 muM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. epiyangambin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 33535454-4 2021 Moreover, eudesmin, fargesin, epimagnolin A, magnolin, and yangambin noncompetitively inhibited UGT1A1-catalyzed SN38 glucuronidation with Ki values of 25.7, 25.3, 3.6, 26.0, and 17.1 muM, respectively, based on kinetic analysis of UGT1A1 inhibition in pooled human liver microsomes. epiyangambin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 232-238 33535454-6 2021 These in vitro results suggest the necessity of evaluating whether the five tetrahydrofurofuranoid lignans can cause drug-drug interactions with UGT1A1 and UGT1A3 substrates in vivo. tetrahydrofurofuranoid 76-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 33535454-6 2021 These in vitro results suggest the necessity of evaluating whether the five tetrahydrofurofuranoid lignans can cause drug-drug interactions with UGT1A1 and UGT1A3 substrates in vivo. Lignans 99-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 33615175-9 2021 Further, formation of glucuronides of raloxifene and 7-ethyl-10-hydroxycamptothecin were reduced in the cells expressing the UGT1A1 P152T mutant. Glucuronides 22-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 33615175-9 2021 Further, formation of glucuronides of raloxifene and 7-ethyl-10-hydroxycamptothecin were reduced in the cells expressing the UGT1A1 P152T mutant. Raloxifene Hydrochloride 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 33615175-9 2021 Further, formation of glucuronides of raloxifene and 7-ethyl-10-hydroxycamptothecin were reduced in the cells expressing the UGT1A1 P152T mutant. Irinotecan 53-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 33551819-6 2020 As expected, CITCO, the direct activator, and PB, the indirect activator of CAR, induced CYP3A4 (31 and 40-fold), CYP2B6 (24 and 28-fold) and UGT1A1 (2.9 and 4.2-fold), respectively. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 33551819-6 2020 As expected, CITCO, the direct activator, and PB, the indirect activator of CAR, induced CYP3A4 (31 and 40-fold), CYP2B6 (24 and 28-fold) and UGT1A1 (2.9 and 4.2-fold), respectively. Phenobarbital 46-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 32648321-12 2020 CONCLUSIONS: CYP1A1 and CYP2C9, UGT1A1, UGT1A7, UGT1A8 and UGT1A9, and MRP4 all played important roles in the metabolism and disposition of bavachin. bavachin 140-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 33154041-2 2021 However, temporal delay in the development of the UGT1A1 gene leads to an accumulation of serum bilirubin in newborn children. Bilirubin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 32813611-0 2021 Main contribution of UGT1A1 and CYP2C9 in the metabolism of UR-1102, a novel agent for the treatment of gout. UR-1102 60-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 32813611-5 2021 Recombinant UGT1A1 and UGT1A3 showed high glucuronidation of UR-1102. UR-1102 61-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 33125947-0 2020 Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 33125947-1 2020 BACKGROUND: Pre-therapeutic UGT1A1 genotyping is not yet routinely performed in most hospitals in patients starting irinotecan chemotherapy. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 33125947-5 2020 A high level of evidence (level I) was found for a higher incidence of irinotecan-induced severe toxicity in homozygous carriers of UGT1A1*28 or UGT1A1*6. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 33125947-5 2020 A high level of evidence (level I) was found for a higher incidence of irinotecan-induced severe toxicity in homozygous carriers of UGT1A1*28 or UGT1A1*6. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 33125947-9 2020 CONCLUSION: Pre-therapeutic genotyping of UGT1A1 in patients initiating treatment with irinotecan improves patient safety, is likely to be cost-saving, and should, therefore, become standard of care. Irinotecan 87-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 33604208-1 2021 Crigler-Najjar syndrome is an inborn error of metabolism caused by a point mutation in one of the five exons of UGT1A1 gene, the product of which is responsible for elimination of bilirubin via bile. Bilirubin 180-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 33387482-3 2021 Treatment of HepaRG cells with 3-deazaadenosine, an inhibitor of RNA methylation, significantly increased UGT1A1, UGT1A3, UGT1A4, UGT1A9, UGT2B7, UGT2B10, and UGT2B15 mRNA levels (1.3- to 2.6-fold). 3-deazaadenosine 31-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 33119477-1 2020 PURPOSE: Differentiating the irinotecan dose on the basis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) genotype improves the pathologic complete response (pCR) rate. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-112 33119477-1 2020 PURPOSE: Differentiating the irinotecan dose on the basis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) genotype improves the pathologic complete response (pCR) rate. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 33119477-14 2020 CONCLUSION: Adding irinotecan guided by UGT1A1 genotype to capecitabine-based neoadjuvant chemoradiotherapy significantly increased complete tumor response in Chinese patients. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 33201683-5 2020 Coexposure of C3A cells to pentachlorophenol (sulfotransferase 1 inhibitor) or ketoconazole (UDP-glucuronosyltransferase 1A inhibitor) potentiated micronuclei induction by each compound, with thresholds lowered from 2.5-5.0 to 0.6-1.2 muM. Ketoconazole 79-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-123 32750744-12 2020 CONCLUSIONS: CYP1A2, 2C19 and UGT1As play an important role in isobavachin metabolism. isobavachin 63-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-34 32750744-8 2020 Additionally, isobavachin underwent efficient oxidation and glucuronidation by human liver microsomes and HIM with CLint values from 5.53 to 148.79 mul/min per mg. CYP1A2, 2C19 contributed 11.3% and 17.1% to hepatic metabolism of isobavachin, respectively, with CLint values from 8.75 to 77.33 mul/min per mg. UGT1As displayed CLint values from 10.73 to 202.62 mul/min per mg for glucuronidation. isobavachin 14-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 310-314 32750744-9 2020 Besides, significant correlation analysis also proved that CYP1A2, 2C19 and UGT1A1, 1A9 were main contributors for the metabolism of isobavachin. isobavachin 133-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-87 32750744-11 2020 Moreover, isobavachin exhibited broad inhibition against CYP2B6, 2C9, 2C19, UGT1A1, 1A9, 2B7 with Ki values from 0.05 to 3.05 mum. isobavachin 10-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 32973940-1 2020 Gilbert syndrome (GS) is a hereditary unconjugated hyperbilirubinemia that results from mutations in the bilirubin uridine diphosphate-glucuronosyltransferase (UGT1A1) gene. Bilirubin 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 32152464-0 2020 UGT1A1 genotype influences clinical outcome in patients with intermediate-risk acute myeloid leukemia treated with cytarabine-based chemotherapy. Cytarabine 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 32737884-0 2020 Pharmacogenetic impact of UGT1A1 polymorphisms on pulmonary neuroendocrine tumours treated with metronomic irinotecan-based chemotherapy in Chinese populations. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 32737884-9 2020 Grade 1 total bilirubin elevation was associated with the homozygous UGT1A1*6 mutation (P = 0.027) or any UGT1A1*6 variants (P = 0.047). Bilirubin 14-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 32737884-9 2020 Grade 1 total bilirubin elevation was associated with the homozygous UGT1A1*6 mutation (P = 0.027) or any UGT1A1*6 variants (P = 0.047). Bilirubin 14-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 32737884-11 2020 CONCLUSIONS: The impact of UGT1A1 polymorphisms varies in different irinotecan-based chemotherapies. Irinotecan 68-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 32737884-12 2020 UGT1A1*6 and UGTA1A*28 were useful for the prediction of irinotecan-related severe toxicity in Chinese PNT patients treated with metronomic IBC. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 32152464-4 2020 Here we report the association between the UGT1A1 rs8175347 genotype and the clinical outcome of 455 intermediate-risk cytogenetic AML patients receiving cytarabine-based chemotherapy. Cytarabine 154-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 32905582-2 2020 In this study, we identified the neuroactive hormone dopamine as an inducer of drug metabolizing enzymes CYP1A1, CYP1B1, and UGT1A1 in colon and glioblastoma cells and similar results were observed for carbidopa. Dopamine 53-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 32985005-5 2020 In this review, the PGx profiles of drug-gene pairs with potential impact in GI malignancy therapy - DPYD-5-fluorouracil/capecitabine and UGT1A1-irinotecan - and the available clinical evidence of their roles in reducing severe adverse events are discussed. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 32936306-0 2020 Association of UGT1A1*6 polymorphism with irinotecan-based chemotherapy reaction in colorectal cancer patients: a systematic review and a meta-analysis. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 32829105-0 2020 Determination of the UGT1A1 polymorphism as guidance for irinotecan dose escalation in metastatic colorectal cancer treated with first-line bevacizumab and FOLFIRI (PURE FIST). Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 32763815-14 2020 Isozymes of CYP1A2, CYP2C9, UGT1A1, UGT1A6, UGT1A9, UGT2B7 and UGT2B15 were involved in phase I and phase II metabolism of osalmid. osalmide 123-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 32250470-11 2020 Aripiprazole, dehydro-aripiprazole and olanzapine pharmacokinetics were significantly affected by polymorphisms in CYP2D6, CYP3A, CYP1A2, ABCB1and UGT1A1 genes. Aripiprazole 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 32250470-11 2020 Aripiprazole, dehydro-aripiprazole and olanzapine pharmacokinetics were significantly affected by polymorphisms in CYP2D6, CYP3A, CYP1A2, ABCB1and UGT1A1 genes. dehydroaripiprazole 14-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 32250470-11 2020 Aripiprazole, dehydro-aripiprazole and olanzapine pharmacokinetics were significantly affected by polymorphisms in CYP2D6, CYP3A, CYP1A2, ABCB1and UGT1A1 genes. Olanzapine 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 32829105-0 2020 Determination of the UGT1A1 polymorphism as guidance for irinotecan dose escalation in metastatic colorectal cancer treated with first-line bevacizumab and FOLFIRI (PURE FIST). FOLFIRI regimen 156-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 32829105-1 2020 AIM: Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility of patients to irinotecan and its toxicity. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-52 32829105-1 2020 AIM: Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility of patients to irinotecan and its toxicity. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 32829105-8 2020 CONCLUSION: The current study revealed that mCRC patients, regardless of KRAS gene status, with UGT1A1 genotyping can tolerate escalated doses of irinotecan and potentially achieve a more favourable clinical outcome without significantly increased toxicities. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 33041817-7 2020 In HepG2 cells, hypoxia, KCZ, and RA inhibited CYP450 isoforms and UGT1A1 expression. Ketoconazole 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 32666389-0 2020 Impact of single-heterozygous UGT1A1 on the clinical outcomes of irinotecan monotherapy after fluoropyrimidine and platinum-based combination therapy for gastric cancer: a multicenter retrospective study. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32666389-0 2020 Impact of single-heterozygous UGT1A1 on the clinical outcomes of irinotecan monotherapy after fluoropyrimidine and platinum-based combination therapy for gastric cancer: a multicenter retrospective study. 2-fluoropyrimidine 94-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32666389-0 2020 Impact of single-heterozygous UGT1A1 on the clinical outcomes of irinotecan monotherapy after fluoropyrimidine and platinum-based combination therapy for gastric cancer: a multicenter retrospective study. Platinum 115-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32666389-2 2020 We investigated the association between clinical outcomes (efficacy and safety) and UGT1A1 status in patients who received irinotecan monotherapy. Irinotecan 123-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 108-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 184-190 33041817-8 2020 Activation of PXR and CAR in cells treated with 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde (CITCO) and rifampicin (Rif) resulted in the enhancement of CYP450 isoforms, UGT1A1, PXR, and CAR. 6-(4-chlorophenyl)-imidazo (2,1-b) thiazole-5-carbaldehyde 48-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 184-190 32778670-3 2020 Thus, the aim of this study was to assess the allele and genotype frequencies and the relationship between CYP3A4/5, DPYD, UGT1A1, ABCB1, and ABCC2 genetic variations and irinotecan-induced toxicity in Thai colorectal cancer patients. Irinotecan 171-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 33005200-4 2020 100 muM of kurarinone strongly inhibited more than 90% of UGT1A1, UGT1A6, CYP1A2, and CYP2C9. kurarinone 11-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 32878631-9 2020 In the MR analysis of the main UGT1A1 SNP (rs6431625), genetically predicted higher levels of total bilirubin were associated with a 7% increase in CRC risk in men (OR = 1.07 (1.02-1.12); P = 0.006; per 1-SD increment of total bilirubin), while there was no association in women (OR = 1.01 (0.96-1.06); P = 0.73). Bilirubin 100-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 32829410-5 2021 Associations were characterized with CYP2B6 and UGT1A1 genotypes that affect efavirenz and INSTI metabolism, respectively. efavirenz 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 32778670-10 2020 This finding suggests that UGT1A1*28, *6, and ABCC2 c.3972C > T might be an important predictor for irinotecan-induced severe neutropenia. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 31495946-5 2020 In patients with complete UGT1A1 deficiency (type 1 CNS [CNS-I]), unconjugated bilirubin levels increase 3-6 mg/dL per day during the newborn period and reach neurologically dangerous levels between 5 and 14 days of age. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 32758288-0 2020 UGT1A1 polymorphism has a prognostic effect in patients with stage IB or II uterine cervical cancer and one or no metastatic pelvic nodes receiving irinotecan chemotherapy: a retrospective study. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-79 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 32361755-0 2020 Evaluation of the effect of UGT1A1 polymorphisms on the pharmacokinetics of oral and long-acting injectable cabotegravir. cabotegravir 108-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 32361755-2 2020 Cabotegravir is primarily metabolized by uridine 5"-diphospho-glucuronosyltransferase (UGT) 1A1, a known polymorphic enzyme with functional variants that can affect drug metabolism and exposure. cabotegravir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-95 32361755-7 2020 A statistically significant (P < 10-5) association between predicted UGT1A1 activity and maximum change in total bilirubin was also observed (2.45-fold asymptomatic increase for low versus normal) without a corresponding change in ALT. Bilirubin 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 32361755-8 2020 CONCLUSIONS: This modest increase in oral and parenteral cabotegravir exposure associated with a reduced function of UGT1A1 is not considered clinically relevant based on accumulated safety data; no dose adjustment is required. cabotegravir 57-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 32265117-12 2020 CONCLUSION: Our analysis suggested higher completion of concurrent irinotecan was associated with better tumor response for patients with locally advanced rectal cancer with UGT1A1*1*1 or UGT1A1*1*28 phenotypes in the neoadjuvant setting, and at least 4 cycles was recommended. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 32265117-12 2020 CONCLUSION: Our analysis suggested higher completion of concurrent irinotecan was associated with better tumor response for patients with locally advanced rectal cancer with UGT1A1*1*1 or UGT1A1*1*28 phenotypes in the neoadjuvant setting, and at least 4 cycles was recommended. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 32179043-4 2020 Since the knockdown of UDP-glucose 6-dehydrogenase, a synthetase of UDPGA, in HEK293 cells stably expressing UGT1A1 (HEK/UGT1A1 cells) resulted in a significant decrease in 4-methylumbelliferone (4-MU) glucuronosyltransferase activity, supplementation of a sufficient amount of UDPGA is required for UGT activity. Hymecromone 173-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 32011683-0 2020 Genetic influence of ABCG2, UGT1A1 and NR1I2 on dolutegravir plasma pharmacokinetics. dolutegravir 48-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 32011683-2 2020 We sought to ascertain the contributory effect of SNPs in four key genes linked to dolutegravir disposition (UGT1A1, ABCG2, CYP3A and NR1I2) on plasma dolutegravir pharmacokinetics. dolutegravir 83-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 32011683-2 2020 We sought to ascertain the contributory effect of SNPs in four key genes linked to dolutegravir disposition (UGT1A1, ABCG2, CYP3A and NR1I2) on plasma dolutegravir pharmacokinetics. dolutegravir 151-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 32011683-8 2020 CONCLUSIONS: This study showed a pharmacogenetic association between dolutegravir pharmacokinetics and variants in the ABCG2, UGT1A1 and NR1I2 genes, particularly when combined. dolutegravir 69-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 32758288-1 2020 BACKGROUND: Uridine diphosphate glucuronosyltransferase 1 family polypeptide A1 (UGT1A1) is a predictive biomarker for the side-effects of irinotecan chemotherapy, which reduces the volume of tumors harboring UGT1A1 polymorphisms. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 32758288-2 2020 We aimed to determine whether UGT1A1 polymorphisms can predict progression-free survival in patients with local cervical cancer treated with irinotecan chemotherapy. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 32758288-10 2020 Among patients with <=1 metastatic lymph node, those with UGT1A1 polymorphisms benefited from irinotecan chemotherapy and demonstrated significantly longer progression-free survival (p = 0.020) than those with wild-type UGT1A1. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 32758288-11 2020 CONCLUSIONS: Irinotecan chemotherapy might be beneficial in patients with cervical cancer, UGT1A1 polymorphisms, and <= 1 metastatic lymph nodes. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 32850329-8 2020 Pharmacogenomic analyses revealed multiple alterations of fluoropyrimidine and irinotecan metabolism: severe deficiency of fluorouracil degradation rate (FUDR), single nucleotide polymorphisms of UGT1A1 *28 variable number of tandem repeats (VNTR) 7R/7R homozygote, ABCB1 c.C3435T, c.C1236T, MTHFR c.C667T homozygote, DPYD c.A166G, TSER 28bp VNTR 2R/3R heterozygote. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 34007623-0 2020 Dosage Adjustment of Irinotecan in Patients with UGT1A1 Polymorphisms: A Review of Current Literature. Irinotecan 21-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 34007623-1 2020 Objective: To review available literature regarding pharmacogenomics (PGx) effects on the metabolism of irinotecan by the UGT1A1 gene and the resulting dose adjustments based on PGx genetic variant. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 34007623-3 2020 The extent of decreased function of UGT1A1 varies based on genotype so irinotecan dose adjustments may be needed. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 34007623-7 2020 Conclusion: These findings add to a growing body of literature that suggest patients with UGT1A1 *28 or *6 variant alleles benefit from lower doses of irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 32612873-1 2020 The human UDP-glucuronosyltransferase 1A1 (UGT1A1), one of the most essential conjugative enzymes, is responsible for the metabolism and detoxification of bilirubin and other endogenous substances, as well as many different xenobiotic compounds. Bilirubin 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-41 32612873-1 2020 The human UDP-glucuronosyltransferase 1A1 (UGT1A1), one of the most essential conjugative enzymes, is responsible for the metabolism and detoxification of bilirubin and other endogenous substances, as well as many different xenobiotic compounds. Bilirubin 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 32264830-0 2020 Correlation between UGT1A1 gene polymorphism and irinotecan chemotherapy in metastatic colorectal cancer: a study from Guangxi Zhuang. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 32264830-14 2020 The UGT1A1 gene polymorphism with irinotecan chemotherapy-associated diarrhea and neutropenia were closely related. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 31980500-3 2020 The expression of UGT1A1 is developmentally delayed in liver and intestines resulting in the accumulation of serum bilirubin during the neonatal period. Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 31233374-10 2020 Glucuronidation of glycyrol was primarily mediated by UGT1A1, UGT1A3, UGT1A9, and UGT2B7. glycyrol 19-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 32323779-0 2020 Sulforaphane suppresses carcinogenesis of colorectal cancer through the ERK/Nrf2-UDP glucuronosyltransferase 1A metabolic axis activation. sulforaphane 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-111 32323779-2 2020 The detoxification of CRC food-borne carcinogenic heterocyclic amines is highly dependent on UDP glucuronosyltransferase 1A (UGT1A)-mediated glucuronidation. Amines 63-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-123 32323779-2 2020 The detoxification of CRC food-borne carcinogenic heterocyclic amines is highly dependent on UDP glucuronosyltransferase 1A (UGT1A)-mediated glucuronidation. Amines 63-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-130 32323779-10 2020 Reverse transcription-quantitative polymerase chain reaction and western blotting were employed to verify the role of Nrf2 in SFN-induced UGT1A. sulforaphane 126-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-143 32323779-17 2020 Pretreatment with PD58059 reversed the SFN-induced subcellular translocation of Nrf2 and the expression of UGT1A. pd58059 18-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-112 30514181-4 2020 Irinotecan is bioactivated into its metabolite SN-38, which is subsequently detoxified by uridine diphosphate-glucuronosyl transferases (mainly UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 30514181-4 2020 Irinotecan is bioactivated into its metabolite SN-38, which is subsequently detoxified by uridine diphosphate-glucuronosyl transferases (mainly UGT1A1). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 31233374-12 2020 In conclusion, glycyrol undergoes the efficient metabolic hydroxylation and glucuronidation reactions in human liver microsomes, which are predominantly catalyzed by CYP1A2, UGT1A1/3/9, and UGT2B7. glycyrol 15-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 31888882-8 2020 After stratification by UGT1A1 phenotypes, there was a significant decrease in total bilirubin among all phenotypes, conjugated bilirubin among intermediate metabolizers, and unconjugated bilirubin among normal and intermediate metabolizers. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 31602632-10 2020 Homozygosity (41%) for the promoter variant of UGT1A1 (Gilbert syndrome) led to a significantly higher mean bilirubin level (126 54 micromol/l) with a higher frequency of cholelithiasis (30%) (P < 0 001). Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 32131538-5 2020 These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic. Maytansine 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 31888882-8 2020 After stratification by UGT1A1 phenotypes, there was a significant decrease in total bilirubin among all phenotypes, conjugated bilirubin among intermediate metabolizers, and unconjugated bilirubin among normal and intermediate metabolizers. Bilirubin 128-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 31888882-8 2020 After stratification by UGT1A1 phenotypes, there was a significant decrease in total bilirubin among all phenotypes, conjugated bilirubin among intermediate metabolizers, and unconjugated bilirubin among normal and intermediate metabolizers. Bilirubin 128-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 31888882-9 2020 The data also show that UGT1A1 genotype predicts serum bilirubin levels at baseline, but this relationship is lost after efavirenz treatment. Bilirubin 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 31888882-11 2020 Our data suggest that efavirenz may alter bilirubin disposition mainly through induction of UGT1A1 metabolism and efflux through multidrug resistance-associated protein 2. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 31812603-4 2020 Our data indicated that PT exhibited potent inhibition against HLM, UGT1A6, UGT1A9, UGT2B7, and UGT2B15, moderate inhibition against UGT1A1, UGT1A3, UGT1A8, and UGT2B4, negligible inhibition against UGT1A4, UGT1A7, UGT1A10, and UGT2B17. pterostilbene 24-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 32058557-2 2020 Evaluation of pharmacogenomically dosed perioperative gFOLFIRINOX (fluorouracil, leucovorin, oxaliplatin, and UGT1A1 genotype-directed irinotecan) to optimize efficacy while limiting toxic effects may have value. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 32204767-2 2020 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), organic anion transporter polypeptide 2 (OATP2), heme oxygenase 1 (HO-1), and biliverdin reductase A (BLVRA) play crucial roles in the metabolism of bilirubin. Bilirubin 207-216 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-47 31691077-4 2020 Our data indicated that sunitinib exhibited competitive inhibition against SN-38 glucuronidation by UGT1A1, but inhibitory effects of sunitinib were weak in pooled human liver microsomes (HLMs) (Ki = 119.00 muM) and recombinant UGT1A1 (Ki = 42.71 muM). sunitinib 24-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 31691077-4 2020 Our data indicated that sunitinib exhibited competitive inhibition against SN-38 glucuronidation by UGT1A1, but inhibitory effects of sunitinib were weak in pooled human liver microsomes (HLMs) (Ki = 119.00 muM) and recombinant UGT1A1 (Ki = 42.71 muM). sunitinib 24-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 228-234 31640912-0 2020 Pazopanib-Induced Liver Toxicity in Patients With Metastatic Renal Cell Carcinoma: Effect of UGT1A1 Polymorphism on Pazopanib Dose Reduction, Safety, and Patient Outcomes. pazopanib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 31640912-11 2020 Patients with UGT1A1 polymorphism safely resumed pazopanib at ultra-low doses determined by the degree of liver toxicity and UGT1A1 polymorphism. pazopanib 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 31640912-11 2020 Patients with UGT1A1 polymorphism safely resumed pazopanib at ultra-low doses determined by the degree of liver toxicity and UGT1A1 polymorphism. pazopanib 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 31640912-13 2020 UGT1A1 assessment could improve the management of pazopanib-induced liver toxicity in patients with mRCC. pazopanib 50-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 31525573-2 2020 The elimination of RAL to RAL glucuronide (RAL GLU) is mediated primarily by UDP glucuronosyltransferase 1A1 (UGT1A1). Glucuronides 30-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-108 32049823-9 2020 Serum TB correlated positively with UGT1A1 mutation load (gamma = 0.281, P = .048), hemoglobin (gamma = .359, P = .010) and hematocrit (gamma = 0.365, P = .010), but negatively with RBC lifespan (gamma = -0.336, P = .017). Bilirubin 6-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 32433020-11 2020 Distinct expression of UGT1A1, CYP2B6, CYP2C9, CYP3A4, and CYP7A1 genes explains the ability to clear toxins and bilirubin as observed in normal hepatocytes. Bilirubin 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 31727359-8 2020 With silymarin, we found that UGT1A1 and, to a lesser extent UGT1A9, were primarily responsible for the glucuronidation of the flavonolignan constituents. Flavonolignans 127-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 31727359-9 2020 It is concluded that the metabolism of silymarin flavonolignans may involve multiple UGT enzymes, of which UGT1A1 appears to play the major role in the glucuronidation. Flavonolignans 39-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 31965023-7 2020 HLSC expressed UGT1A1 in vivo, induced a strong decrease in serum unconjugated bilirubin, thus significantly improving phenotype and survival compared to untreated controls. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 31525573-2 2020 The elimination of RAL to RAL glucuronide (RAL GLU) is mediated primarily by UDP glucuronosyltransferase 1A1 (UGT1A1). Glucuronides 30-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 31525573-2 2020 The elimination of RAL to RAL glucuronide (RAL GLU) is mediated primarily by UDP glucuronosyltransferase 1A1 (UGT1A1). Glutamic Acid 47-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-108 31525573-2 2020 The elimination of RAL to RAL glucuronide (RAL GLU) is mediated primarily by UDP glucuronosyltransferase 1A1 (UGT1A1). Glutamic Acid 47-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 31740989-5 2020 In agreement with our previous genotyping data, the genetic polymorphism of uridine 5"-diphospho-glucuronosyltransferase (UGT)1A1*6, which affects the glucuronidation activity and circulating glucuronic acid level was identified as one of the determinants of MMI-induced hepatotoxicity. Uridine 76-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 31740989-5 2020 In agreement with our previous genotyping data, the genetic polymorphism of uridine 5"-diphospho-glucuronosyltransferase (UGT)1A1*6, which affects the glucuronidation activity and circulating glucuronic acid level was identified as one of the determinants of MMI-induced hepatotoxicity. Glucuronic Acid 192-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 32237326-1 2020 The bilirubin metabolism mediated by the phase II metabolizing enzyme UGT1A1 in the liver was evaluated to study the potential hepatotoxicity risk based on investigation on the inhibitory effect of rhein and its metabolites on the UGT1A1 enzyme in Rhei Radix et Rhizoma. Bilirubin 4-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 31707444-8 2020 CONCLUSIONS: The disposition of PEGylated SN-38 is independent of UGT1A activity in rats, and PLX038 may find utility in full-dose treatment of patients who are UGT1A1*28 homozygotes or have metastatic disease with coincidental or incidental liver dysfunction. 5-(1H-pyrrolo(2,3-b)pyridin-3-ylmethyl)-N-((4-(trifluoromethyl)phenyl)methyl)pyridin-2-amine 94-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 33622990-2 2020 This polymorphism leads to 30% of normal rate transcription initiation of UGT1A1 gene, thus decreasing the bilirubin glucuronidation. Bilirubin 107-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 31342573-6 2020 The transplanted pHSCs underwent maturation in vivo to restore the deficient metabolic hepatic function (bilirubin glucuronidation by UGT1A1). Bilirubin 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 31558477-0 2020 Clinical Assessment of 5-Fluorouracil/Leucovorin, Nab-Paclitaxel, and Irinotecan (FOLFIRABRAX) in Untreated Patients with Gastrointestinal Cancer Using UGT1A1 Genotype-Guided Dosing. Fluorouracil 23-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 31558477-0 2020 Clinical Assessment of 5-Fluorouracil/Leucovorin, Nab-Paclitaxel, and Irinotecan (FOLFIRABRAX) in Untreated Patients with Gastrointestinal Cancer Using UGT1A1 Genotype-Guided Dosing. folfirabrax 82-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 48-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 48-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 31558477-2 2020 UDP Glucuronosyltransferase 1A1 (UGT1A1) clears SN-38, the active metabolite of irinotecan. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 31558477-4 2020 We performed a trial to assess the safety and tolerability of FOLFIRABRAX with UGT1A1 genotype-guided dosing of irinotecan. folfirabrax 62-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 31558477-4 2020 We performed a trial to assess the safety and tolerability of FOLFIRABRAX with UGT1A1 genotype-guided dosing of irinotecan. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. folfirabrax 13-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. folfirabrax 13-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 31558477-16 2020 CONCLUSIONS: FOLFIRABRAX with genotype-guided dosing of irinotecan is tolerable in patients with advanced gastrointestinal cancer and UGT1A1*1*1 or UGT1A1*1*28 genotypes. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 31254178-0 2019 Effects of UGT1A1 Polymorphism, Gender and Triglyceride on the Pharmacokinetics of Telmisartan in Chinese Patients with Hypertension: A Population Pharmacokinetic Analysis. Telmisartan 83-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 31254178-3 2019 In this study, we aimed to develop a population pharmacokinetic (PopPK) model to quantify the effects of gender and UGT1A1 polymorphisms on the pharmacokinetics of telmisartan. Telmisartan 164-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 31254178-9 2019 UGT1A1 (rs4124874) affected the bioavailability (F1) of telmisartan. Telmisartan 56-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 31254178-11 2019 CONCLUSION: A PopPK model of telmisartan was established to confirm that UGT1A1 genotype, gender and triglyceride can affect the pharmacokinetics of telmisartan in Chinese patients with hypertension. Telmisartan 29-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 31254178-11 2019 CONCLUSION: A PopPK model of telmisartan was established to confirm that UGT1A1 genotype, gender and triglyceride can affect the pharmacokinetics of telmisartan in Chinese patients with hypertension. Telmisartan 149-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 31586129-1 2019 It has been reported that there are differences in effects on irinotecan-induced adverse reactions between UGT1A1*6 and UGT1A1*28. irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 31586129-1 2019 It has been reported that there are differences in effects on irinotecan-induced adverse reactions between UGT1A1*6 and UGT1A1*28. irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 31586129-2 2019 In order to compare those differences in the Japanese population, we examined the associations between UGT1A1 and irinotecan-induced adverse reactions using the BioBank Japan Project database. irinotecan 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 31730632-2 2019 For example, adverse drug reactions when patients receive CPT-11 (irinotecan) such as in cancer chemotherapy are caused by amino acid substitutions in UGT1A1. irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 31621310-2 2019 Therefore, we examined the structure-activity relationships of isoflavones and isomeric flavone and flavanones as AhR ligands on the basis of their induction of CYP1A1, CYP1B1, and UGT1A1 gene expression in colon cancer Caco2 cells and young adult mouse colonocyte (YAMC) cells. Isoflavones 63-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 31621310-2 2019 Therefore, we examined the structure-activity relationships of isoflavones and isomeric flavone and flavanones as AhR ligands on the basis of their induction of CYP1A1, CYP1B1, and UGT1A1 gene expression in colon cancer Caco2 cells and young adult mouse colonocyte (YAMC) cells. Flavanones 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 31621310-5 2019 For example, 4",5,7-trihydroxyisoflavone (genistein) was AhR-inactive whereas 4",5,7-trihydroxyflavone (apigenin) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells. 5,7,4'-trihydroxyflavone 78-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 31621310-5 2019 For example, 4",5,7-trihydroxyisoflavone (genistein) was AhR-inactive whereas 4",5,7-trihydroxyflavone (apigenin) induced CYP1A1, CYP1B1, and UGT1A1 in Caco2 cells. Apigenin 104-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 31730632-5 2019 We then derived a mathematical model that can predict the conjugation capacities of mutant UGT1A1s by using results of substrate docking in silico and results of in vitro analysis of glucuronidation of acetaminophen and 17beta-estradiol by UGT1A1s. Acetaminophen 202-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 31730632-5 2019 We then derived a mathematical model that can predict the conjugation capacities of mutant UGT1A1s by using results of substrate docking in silico and results of in vitro analysis of glucuronidation of acetaminophen and 17beta-estradiol by UGT1A1s. Acetaminophen 202-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 240-246 31730632-5 2019 We then derived a mathematical model that can predict the conjugation capacities of mutant UGT1A1s by using results of substrate docking in silico and results of in vitro analysis of glucuronidation of acetaminophen and 17beta-estradiol by UGT1A1s. Estradiol 220-236 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 31730632-6 2019 This experimental procedure showed that the in silico conjugation capacities of other mutant UGT1A1s with bilirubin or SN-38 were similar to reported in vitro conjugation capacities. Bilirubin 106-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 31715674-8 2019 Conclusions: The related gene UGT1A1 is related to the metabolism of thyroxine in the liver. Thyroxine 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 31715674-11 2019 Abnormal serum thyroxine levels during pregnancy may be associated with abnormal metabolism of thyroxine caused by UGT1A1 mutation. Thyroxine 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 31715674-11 2019 Abnormal serum thyroxine levels during pregnancy may be associated with abnormal metabolism of thyroxine caused by UGT1A1 mutation. Thyroxine 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 31445021-6 2019 The structures of dabrafenib, ibrutinib, lapatinib, nintedanib, pazopanib, regorafenib, trametinib and 22 other KIs overlaid well on that of sorafenib, a potent inhibitor of UGT1A1 and UGTs 1A7-1A10. Sorafenib 141-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 31840682-6 2019 Thus, potent inducers of UGT1A1 and CYP3A4 (e.g., rifamycins/anticonvulsants) should be avoided due to significantly decreased BIC serum exposure. rifamycin S 50-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 31445021-1 2019 We have demonstrated previously that the kinase inhibitors (KIs) lapatinib, pazopanib, regorafenib and sorafenib are potent inhibitors of UGT1A1 and UGTs 1A7-1A10. Lapatinib 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 31445021-1 2019 We have demonstrated previously that the kinase inhibitors (KIs) lapatinib, pazopanib, regorafenib and sorafenib are potent inhibitors of UGT1A1 and UGTs 1A7-1A10. pazopanib 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 31445021-1 2019 We have demonstrated previously that the kinase inhibitors (KIs) lapatinib, pazopanib, regorafenib and sorafenib are potent inhibitors of UGT1A1 and UGTs 1A7-1A10. regorafenib 87-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 31445021-1 2019 We have demonstrated previously that the kinase inhibitors (KIs) lapatinib, pazopanib, regorafenib and sorafenib are potent inhibitors of UGT1A1 and UGTs 1A7-1A10. Sorafenib 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 31445021-3 2019 Dabrafenib, ibrutinib, nintedanib and trametinib were potent inhibitors of human liver microsomal UGT1A1; Ki,u values ranged from 1.1 to 7.5 microM. dabrafenib 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 31445021-3 2019 Dabrafenib, ibrutinib, nintedanib and trametinib were potent inhibitors of human liver microsomal UGT1A1; Ki,u values ranged from 1.1 to 7.5 microM. ibrutinib 12-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 31445021-3 2019 Dabrafenib, ibrutinib, nintedanib and trametinib were potent inhibitors of human liver microsomal UGT1A1; Ki,u values ranged from 1.1 to 7.5 microM. nintedanib 23-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 31445021-3 2019 Dabrafenib, ibrutinib, nintedanib and trametinib were potent inhibitors of human liver microsomal UGT1A1; Ki,u values ranged from 1.1 to 7.5 microM. trametinib 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 31840682-6 2019 Thus, potent inducers of UGT1A1 and CYP3A4 (e.g., rifamycins/anticonvulsants) should be avoided due to significantly decreased BIC serum exposure. bictegravir 127-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 31207516-8 2019 At concentrations relevant for human exposure (ng/mL range) BPA and its analogues as individual compounds and in mixtures did not exert genotoxic activity, whereas BPA and BPAF as well as the mixtures up-regulated the expressions of CYP1A1 and UGT1A1. bisphenol A 164-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 244-250 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorooctanoic acid 213-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorooctanoic acid 237-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorododecanoic acid 247-271 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorododecanoic acid 273-278 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-11 2019 PFDA and PFOS exhibited competitive inhibition towards UGT1A1, and PFDA and PFTA showed competitive inhibition towards UGT1A8. perfluorodecanoic acid 0-4 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 31472452-11 2019 PFDA and PFOS exhibited competitive inhibition towards UGT1A1, and PFDA and PFTA showed competitive inhibition towards UGT1A8. perfluorooctane sulfonic acid 9-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 31472452-13 2019 The values were calculated to be 0.3 mumoL/L and 1.3 mumoL/L for the in vivo inhibition of PFDA towards UGT1A1-and UGT1A8-catalyzed metabolism of substances, and 0.2 mumoL/L and 2.0 mumoL/L for the inhibition of PFOS towards UGT1A1 and the inhibition of PFTA towards UGT1A8, respectively. perfluorodecanoic acid 91-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 31472452-13 2019 The values were calculated to be 0.3 mumoL/L and 1.3 mumoL/L for the in vivo inhibition of PFDA towards UGT1A1-and UGT1A8-catalyzed metabolism of substances, and 0.2 mumoL/L and 2.0 mumoL/L for the inhibition of PFOS towards UGT1A1 and the inhibition of PFTA towards UGT1A8, respectively. perfluorodecanoic acid 91-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 225-231 31554401-5 2019 (2R)-8-PN was a better substrate for all 11 UGTs except for UGT1A1, which formed more of both (2S)-8-PN glucuronides than (2R)-8-PN glucuronides. 8-prenylnaringenin 94-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 31554401-5 2019 (2R)-8-PN was a better substrate for all 11 UGTs except for UGT1A1, which formed more of both (2S)-8-PN glucuronides than (2R)-8-PN glucuronides. 8-prenylnaringenin 122-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorodecanoic acid 94-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorodecanoic acid 118-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. Potassium perfluorooctanesulfonate 125-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. perfluorooctane sulfonic acid 170-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. Perfluorotetradecanoic acid 177-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31472452-7 2019 The activity of UGT1A1 was inhibited by 98.8%, 98%, 79.9%, 77.1%, and 76.9% at 100 mumoL/L of perfluorodecanoic acid (PFDA), perfluorooctanesulfonic acid potassium salt (PFOS), perfluorotetradecanoic acid (PFTA), perfluorooctanoic acid (PFOA) and perfluorododecanoic acid (PFDoA), respectively. pifithrin 206-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 31619193-0 2019 Uridine diphosphate glucuronosyl transferase 1A (UGT1A1) promoter polymorphism in young patients with sickle cell anaemia: report of the first cohort study from Nigeria. Uridine Diphosphate 0-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 31619193-1 2019 BACKGROUND: (TA) n repeat sequence (rs8175347) of UGT1A1 gene promoter polymorphism is associated with serum bilirubin levels and gallstones among different sickle cell anaemia (SCA) populations. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 31619193-13 2019 Although, bilirubin and fetal hemoglobin (HbF) of patients with gallstones were significantly different from those without gallstone, only the serum bilirubin was associated with UGT1A1 (TA) n genotypes on multivariate analysis (p < 0.0001). Bilirubin 149-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 31207516-8 2019 At concentrations relevant for human exposure (ng/mL range) BPA and its analogues as individual compounds and in mixtures did not exert genotoxic activity, whereas BPA and BPAF as well as the mixtures up-regulated the expressions of CYP1A1 and UGT1A1. 4,4'-hexafluorisopropylidene diphenol 172-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 244-250 30484368-6 2019 The half-maximal inhibitory concentration (IC50) of PTL on the activity of UGT1A1 was determined to be 64.4 muM. parthenolide 52-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 31649539-11 2019 Similarities in allele frequency, comparing major continental groups for VIP pharmacogenes, hint towards a comparable PGx for drugs metabolized by UGT1A1, DPYD, ABCB1, CBR3, COMT, and TPMT; in contrast to variants on CYP3A5 and CYP2B6 for which significant MAF differences were identified. Epoprostenol 118-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 30484368-7 2019 Inhibition kinetics determination showed that PTL exerted noncompetitive inhibition toward UGT1A1, and the inhibition kinetic constant (Ki) was determined to be 12.1 muM. parthenolide 46-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 30484368-8 2019 In silico docking method has been employed to show that hydrogen bonds between PTL and the activity cavity of UGT1A1 contributed to the stronger inhibition of PTL on the activity of UGT1A1 than MCL. Hydrogen 56-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 30484368-8 2019 In silico docking method has been employed to show that hydrogen bonds between PTL and the activity cavity of UGT1A1 contributed to the stronger inhibition of PTL on the activity of UGT1A1 than MCL. Hydrogen 56-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 30484368-8 2019 In silico docking method has been employed to show that hydrogen bonds between PTL and the activity cavity of UGT1A1 contributed to the stronger inhibition of PTL on the activity of UGT1A1 than MCL. parthenolide 79-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 30484368-8 2019 In silico docking method has been employed to show that hydrogen bonds between PTL and the activity cavity of UGT1A1 contributed to the stronger inhibition of PTL on the activity of UGT1A1 than MCL. parthenolide 79-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 30484368-9 2019 In conclusion, PTL can more easily induce drug-drug interaction (DDI) with clinical drugs mainly undergoing UGT1A1-catalyzed glucuronidation. parthenolide 15-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 31128210-6 2019 The further enzyme kinetic studies demonstrated that shikonin was not only a competitive inhibitor of human UGT1A1, UGT1A9, and UGT2B7, but also presented competitive inhibition on rat UGT1A1 and AZTG reactions. shikonin 53-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 31611795-0 2019 Oleanolic Acid and Ursolic Acid Induce UGT1A1 Expression in HepG2 Cells by Activating PXR Rather Than CAR. Oleanolic Acid 0-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 31611795-0 2019 Oleanolic Acid and Ursolic Acid Induce UGT1A1 Expression in HepG2 Cells by Activating PXR Rather Than CAR. ursolic acid 19-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 31611795-6 2019 Results: In HepG2 cells, OA and UA both significantly induced the expression of UGT1A1, UGT1A3, UGT1A4, and UGT1A9 and upregulated the expression of PXR. Oleanolic Acid 25-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 31611795-6 2019 Results: In HepG2 cells, OA and UA both significantly induced the expression of UGT1A1, UGT1A3, UGT1A4, and UGT1A9 and upregulated the expression of PXR. ursolic acid 32-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 31611795-8 2019 A dual-luciferase reporter assay showed that OA and UA could markedly promote PXR-mediated UGT1A1 luciferase activity, whereas OA and UA did not affect CAR-mediated UGT1A1 luciferase activity. Oleanolic Acid 45-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 31611795-8 2019 A dual-luciferase reporter assay showed that OA and UA could markedly promote PXR-mediated UGT1A1 luciferase activity, whereas OA and UA did not affect CAR-mediated UGT1A1 luciferase activity. ursolic acid 52-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 31611795-10 2019 However, the expression of UGT1A1 in hCAR-silenced HepG2 cells was markedly elevated compared to the control group or with non-silenced HepG2 cells treated with OA (10, 20, and 40 muM) or UA (10, 20, and 40 muM). Oleanolic Acid 161-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 31611795-10 2019 However, the expression of UGT1A1 in hCAR-silenced HepG2 cells was markedly elevated compared to the control group or with non-silenced HepG2 cells treated with OA (10, 20, and 40 muM) or UA (10, 20, and 40 muM). ursolic acid 188-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 31611795-11 2019 Conclusions: OA and UA significantly induce the expression of UGT1A1, UGT1A3, UGT1A4, and UGT1A9 in HepG2 cells, and their induction on UGT1A1 is mediated by PXR activation, not CAR. Oleanolic Acid 13-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 31611795-11 2019 Conclusions: OA and UA significantly induce the expression of UGT1A1, UGT1A3, UGT1A4, and UGT1A9 in HepG2 cells, and their induction on UGT1A1 is mediated by PXR activation, not CAR. ursolic acid 20-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 31022310-0 2019 Pharmacogenetic Analysis of OATP1B1, UGT1A1, and BCRP Variants in Relation to the Pharmacokinetics of Letermovir in Previously Conducted Clinical Studies. letermovir 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 30588615-3 2019 Uridine 5"-diphospho-glucuronosyl transferase (UGT)1A1 variants coding for bilirubin UDP-glucuronosyl transferase resulting in mild hyperbilirubinemia (as in Gilbert syndrome (GS)) may confer a strong genetic advantage. Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-54 31022310-2 2019 In this analysis, functional variants in solute carrier organic anion transporter family member 1B1 (SLCO1B1), uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1), and breast cancer resistance protein (BCRP) were assessed for effects on letermovir pharmacokinetics (PK) using pooled genetic information from 296 participants in 12 phase 1 studies. letermovir 242-252 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-158 31022310-5 2019 The UGT1A1*6 variant rs4148323 A allele was present predominantly in Asian participants and was associated with an increase in letermovir AUC compared with noncarriers (GMR, 1.36; 95%CI, 1. letermovir 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 31872743-9 2019 The results of two experiments showed that apigenin was the substrate of UGT1 A1 enzyme,which could inhibit the activity of UGT1 A1 enzyme competitively,and there was a risk of drug interaction between apigenin and UGT1 A1 enzyme substrate in clinical use. Apigenin 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-80 31872743-1 2019 The purpose of this study was to investigate the effect of apigenin on UGT1 A1 enzyme activity and to predict the potential drug-drug interaction of apigenin in clinical use. Apigenin 59-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-78 31872743-2 2019 First,on the basis of previous experiments,the binding targets and binding strength of apigenin to UGT1 A1 enzyme were predicted by computer molecular docking method. Apigenin 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-106 31872743-9 2019 The results of two experiments showed that apigenin was the substrate of UGT1 A1 enzyme,which could inhibit the activity of UGT1 A1 enzyme competitively,and there was a risk of drug interaction between apigenin and UGT1 A1 enzyme substrate in clinical use. Apigenin 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-131 31872743-9 2019 The results of two experiments showed that apigenin was the substrate of UGT1 A1 enzyme,which could inhibit the activity of UGT1 A1 enzyme competitively,and there was a risk of drug interaction between apigenin and UGT1 A1 enzyme substrate in clinical use. Apigenin 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-131 31339084-0 2021 UGT1A1 mutations and psychoses: towards understanding the relationship with unconjugated bilirubin. Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 31872743-9 2019 The results of two experiments showed that apigenin was the substrate of UGT1 A1 enzyme,which could inhibit the activity of UGT1 A1 enzyme competitively,and there was a risk of drug interaction between apigenin and UGT1 A1 enzyme substrate in clinical use. Apigenin 202-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-80 31872743-3 2019 Then the inhibitory effect of apigenin on UGT1 A1 enzyme was evaluated by in vitro human liver microsomal incubation system. Apigenin 30-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-49 31872743-4 2019 Molecular docking results showed that apigenin was docked into the active region of UGT1 A1 enzyme protein F,consistent with the active region of bilirubin docking,with moderate affinity. Apigenin 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-91 31872743-4 2019 Molecular docking results showed that apigenin was docked into the active region of UGT1 A1 enzyme protein F,consistent with the active region of bilirubin docking,with moderate affinity. Bilirubin 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-91 31872743-7 2019 These results suggested that the flavonoid mother nucleus structure had a special structure binding to the enzyme protein UGT1 A1,and the introduction of hydroxyl groups into the mother nucleus can increase the binding ability. Flavonoids 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 31872743-7 2019 These results suggested that the flavonoid mother nucleus structure had a special structure binding to the enzyme protein UGT1 A1,and the introduction of hydroxyl groups into the mother nucleus can increase the binding ability. Hydroxyl Radical 154-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 31872743-8 2019 In vitro inhibition experiments showed that apigenin had a moderate inhibitory effect on UGT1 A1 enzyme in a way of competitive inhibition,which was consistent with the results of molecular docking. Apigenin 44-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-96 30820844-8 2019 UGT1A1, 1A3, 1A6, 1A8, 1A9 and 1A10 participated in the formation of D-luciferin glucuronide, with UGT1A1 exhibiting the highest catalytic activity. d-luciferin glucuronide 69-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30820844-8 2019 UGT1A1, 1A3, 1A6, 1A8, 1A9 and 1A10 participated in the formation of D-luciferin glucuronide, with UGT1A1 exhibiting the highest catalytic activity. d-luciferin glucuronide 69-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 30820844-9 2019 Both chemical inhibition assays and kinetic analysis showed that UGT1A1 and UGT1A3 played important roles in D-luciferin-6"-O-glucuronidation in HLM and HIM, with UGT1A6 also giving a non-negligible contribution to this biotransformation in HLM. D-luciferin 109-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30820844-9 2019 Both chemical inhibition assays and kinetic analysis showed that UGT1A1 and UGT1A3 played important roles in D-luciferin-6"-O-glucuronidation in HLM and HIM, with UGT1A6 also giving a non-negligible contribution to this biotransformation in HLM. 6"-o 121-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30820844-10 2019 CONCLUSIONS: UGT1A1, UGT1A3 and UGT1A6 were responsible for 6"-O-glucuronidation of D-luciferin in HLM, while UGT1A1 and UGT1A3 were the major contributors to this biotransformation in HIM. 6"-o 60-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 30820844-10 2019 CONCLUSIONS: UGT1A1, UGT1A3 and UGT1A6 were responsible for 6"-O-glucuronidation of D-luciferin in HLM, while UGT1A1 and UGT1A3 were the major contributors to this biotransformation in HIM. D-luciferin 84-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 31175220-1 2019 Cabotegravir, a novel integrase inhibitor under development for treatment and prevention of HIV, is primarily metabolized by UDP-glucuronosyltransferase (UGT)1A1 and UGT1A9 to a direct ether glucuronide metabolite. cabotegravir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-161 31175220-1 2019 Cabotegravir, a novel integrase inhibitor under development for treatment and prevention of HIV, is primarily metabolized by UDP-glucuronosyltransferase (UGT)1A1 and UGT1A9 to a direct ether glucuronide metabolite. ether glucuronide 185-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-161 31207142-6 2019 The family based evaluation of the variants showed that the variant, c.714T > G (p.Tyr238Ter), in the CLEC7A gene in first family; the variant, c.55C > T (p.Arg19Ter), in the APOC3 gene and the variant, c.1171C > T (p.Gln391Ter), in the MUTYH gene in second family; and the variant, c.211G > A (p.Gly71Arg), in the UGT1A1 gene in the third family, were found statistically significant (p < 0.05). arg19ter 157-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 315-321 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 30603911-0 2019 Association between UGT1A1 gene polymorphism and safety and efficacy of irinotecan monotherapy as the third-line treatment for advanced gastric cancer. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 30603911-1 2019 BACKGROUND: While uridine diphosphate glucuronosyltransferase (UGT) 1A1 is a key enzyme in the metabolism of irinotecan, relationship between UGT1A1 genotype and safety and efficacy of irinotecan monotherapy in patients with advanced gastric cancer is not clarified. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-71 30603911-1 2019 BACKGROUND: While uridine diphosphate glucuronosyltransferase (UGT) 1A1 is a key enzyme in the metabolism of irinotecan, relationship between UGT1A1 genotype and safety and efficacy of irinotecan monotherapy in patients with advanced gastric cancer is not clarified. Irinotecan 185-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 30603911-8 2019 CONCLUSIONS: The UGT1A1 polymorphism may be related to the clinical outcomes of irinotecan monotherapy as the third-line treatment for advanced gastric cancer. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 31017737-7 2019 Among the 178 patients of Gilbert"s syndrome, serum bilirubin value was inversely correlated with UGT1A1 activity (r = -.306, P < .001). Bilirubin 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 31017737-9 2019 Our results demonstrate that UGT1A1 activity is certainly a determinate for serum bilirubin value and UGT1A1 activity 40% of normal is a proper risk factor for the development of Gilbert"s syndrome. Bilirubin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 31217818-0 2019 An expansion study of genotype-driven weekly irinotecan and capecitabine in combination with neoadjuvant radiotherapy for locally advanced rectal cancer with UGT1A1 *1*1 genotype. Irinotecan 45-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 31217818-0 2019 An expansion study of genotype-driven weekly irinotecan and capecitabine in combination with neoadjuvant radiotherapy for locally advanced rectal cancer with UGT1A1 *1*1 genotype. Capecitabine 60-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 31217818-1 2019 Background: In our previous dose-escalation study, we uncovered the maximum tolerated dose (MTD) of weekly irinotecan was escalated to 80 mg/m2 and 65 mg/m2 for UDP glucuronosyltransferase family 1 member A1 (UGT1A1) *1*1 and *1*28 rectal cancer patients in neoadjuvant chemoradiotherapy (nCRT). Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-207 31217818-1 2019 Background: In our previous dose-escalation study, we uncovered the maximum tolerated dose (MTD) of weekly irinotecan was escalated to 80 mg/m2 and 65 mg/m2 for UDP glucuronosyltransferase family 1 member A1 (UGT1A1) *1*1 and *1*28 rectal cancer patients in neoadjuvant chemoradiotherapy (nCRT). Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 30968312-9 2019 For illustration purpose, the approaches were also applied to an irinotecan mixture model demonstrating 36% lower clearance of irinotecan metabolite (SN-38) in individuals with UGT1A1 homo/heterozygote versus wild-type genotype. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 31150474-0 2019 Mechanism of the efflux transport of demethoxycurcumin-O-glucuronides in HeLa cells stably transfected with UDP-glucuronosyltransferase 1A1. demethoxycurcumin-o-glucuronides 37-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-139 30044687-7 2019 UGT1A1, UGT1A3, UGT1A7, UGT1A8, UGT1A9, UGT1A10 and UGT2B7 participated in the formation of 4-O-glucuronide, with UGT1A9 exhibiting the highest catalytic activity in this biotransformation. 4-o-glucuronide 92-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30044687-8 2019 Only UGT1A1 and UGT1A3 were involved in the formation of 4"-O-glucuronide, exhibiting similar reaction rates. 4"-o-glucuronide 57-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 30044687-10 2019 UGT1A9 was the major contributor to the formation of LCA-4-O-glucuronide, while UGT1A1 played important roles in both LCA-4-O- and 4"-O-glucuronidation. 4"-o 131-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 31150474-3 2019 In this study, we aimed to investigate the formation of DMC-O-glucuronides by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) and its transport by breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRPs) in HeLa cells stably transfected with UGT1A1 (named HeLa1A1 cells). dmc-o-glucuronides 56-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-126 31150474-3 2019 In this study, we aimed to investigate the formation of DMC-O-glucuronides by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) and its transport by breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRPs) in HeLa cells stably transfected with UGT1A1 (named HeLa1A1 cells). dmc-o-glucuronides 56-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 287-293 31150474-3 2019 In this study, we aimed to investigate the formation of DMC-O-glucuronides by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) and its transport by breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRPs) in HeLa cells stably transfected with UGT1A1 (named HeLa1A1 cells). dmc-o-glucuronides 56-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 30645764-0 2019 A UGT1A1 genotype-guided dosing study of modified FOLFIRINOX in previously untreated patients with advanced gastrointestinal malignancies. folfirinox 50-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 31142299-2 2019 CNs type II patients usually benefit from phenobarbital treatment that induces residual UGT1A1 activity. Phenobarbital 42-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 30645764-2 2019 UGT1A1 (UDP glucuronosyltransferase 1A1) eliminates the active metabolite of irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30645764-2 2019 UGT1A1 (UDP glucuronosyltransferase 1A1) eliminates the active metabolite of irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 30645764-4 2019 The primary objective was to determine the dose-limiting toxicity (DLT) rate in cycle 1 of modified FOLFIRINOX (mFOLFIRINOX) using genotype-guided dosing of irinotecan for the most common UGT1A1 genotypes (*1/*1, *1/*28) in advanced gastrointestinal malignancies, with expansion in pancreatic and biliary tract cancers. folfirinox 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 30645764-4 2019 The primary objective was to determine the dose-limiting toxicity (DLT) rate in cycle 1 of modified FOLFIRINOX (mFOLFIRINOX) using genotype-guided dosing of irinotecan for the most common UGT1A1 genotypes (*1/*1, *1/*28) in advanced gastrointestinal malignancies, with expansion in pancreatic and biliary tract cancers. mfolfirinox 112-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 30645764-6 2019 Irinotecan doses of 180, 135, and 90 mg/m2 were administered for UGT1A1 genotypes *1/*1, *1/*28, and *28/*28, respectively. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30645764-13 2019 CONCLUSION: On the basis of our prespecified criteria, tolerability of UGT1A1 genotype-guided mFOLFIRINOX was not established in pancreatic and biliary tract cancers. mfolfirinox 94-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 30962262-0 2019 Association between the UGT1A1*28 allele and hyperbilirubinemia in HIV-positive patients receiving atazanavir: a meta-analysis. Atazanavir Sulfate 99-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 30962262-1 2019 Objectives The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1)*28 allele in HIV-positive patients receiving atazanavir (ATV) might be associated with the risk of hyperbilirubinemia. Atazanavir Sulfate 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-62 30962262-1 2019 Objectives The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1)*28 allele in HIV-positive patients receiving atazanavir (ATV) might be associated with the risk of hyperbilirubinemia. Atazanavir Sulfate 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 30962262-1 2019 Objectives The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1)*28 allele in HIV-positive patients receiving atazanavir (ATV) might be associated with the risk of hyperbilirubinemia. Atazanavir Sulfate 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-62 30962262-1 2019 Objectives The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1)*28 allele in HIV-positive patients receiving atazanavir (ATV) might be associated with the risk of hyperbilirubinemia. Atazanavir Sulfate 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 30962262-7 2019 A significantly increased risk of hyperbilirubinemia was observed in HIV-positive patients receiving ATV with the UGT1A1*1/*28 or UGT1A1*28/*28 genotype, and the risk was higher with the UGT1A1*28/*28 genotype than with the UGT1A1*1/*28 genotype. Atazanavir Sulfate 101-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 30962262-7 2019 A significantly increased risk of hyperbilirubinemia was observed in HIV-positive patients receiving ATV with the UGT1A1*1/*28 or UGT1A1*28/*28 genotype, and the risk was higher with the UGT1A1*28/*28 genotype than with the UGT1A1*1/*28 genotype. Atazanavir Sulfate 101-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 30962262-7 2019 A significantly increased risk of hyperbilirubinemia was observed in HIV-positive patients receiving ATV with the UGT1A1*1/*28 or UGT1A1*28/*28 genotype, and the risk was higher with the UGT1A1*28/*28 genotype than with the UGT1A1*1/*28 genotype. Atazanavir Sulfate 101-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 30962262-7 2019 A significantly increased risk of hyperbilirubinemia was observed in HIV-positive patients receiving ATV with the UGT1A1*1/*28 or UGT1A1*28/*28 genotype, and the risk was higher with the UGT1A1*28/*28 genotype than with the UGT1A1*1/*28 genotype. Atazanavir Sulfate 101-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 30962262-9 2019 All of the pooled ORs were not significantly affected by the remaining studies and different modeling methods, indicating robust results.Conclusions This meta-analysis suggests that the UGT1A1*28 allele represents a biomarker for an increased risk of hyperbilirubinemia in HIV-positive patients receiving ATV. Atazanavir Sulfate 305-308 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 30589990-4 2019 Studies verified predisposition of HIV-positive carriers of UGT1A1*28 to severe atazanavir-induced hyperbilirubinaemia, intolerance to 5-fluorouracyl in gastrointestinal cancer patients with deleterious DPYD variants, failure of HCV-infected carriers of IFNL3 rs12979860 to obtain a sustained viral response to PEG-IFN-alpha, and hypersensitivity reactions to abacavir in HIV-positive carriers of HLA-B*57:01. Atazanavir Sulfate 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 30589990-4 2019 Studies verified predisposition of HIV-positive carriers of UGT1A1*28 to severe atazanavir-induced hyperbilirubinaemia, intolerance to 5-fluorouracyl in gastrointestinal cancer patients with deleterious DPYD variants, failure of HCV-infected carriers of IFNL3 rs12979860 to obtain a sustained viral response to PEG-IFN-alpha, and hypersensitivity reactions to abacavir in HIV-positive carriers of HLA-B*57:01. 5-fluorouracyl 135-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 30589990-4 2019 Studies verified predisposition of HIV-positive carriers of UGT1A1*28 to severe atazanavir-induced hyperbilirubinaemia, intolerance to 5-fluorouracyl in gastrointestinal cancer patients with deleterious DPYD variants, failure of HCV-infected carriers of IFNL3 rs12979860 to obtain a sustained viral response to PEG-IFN-alpha, and hypersensitivity reactions to abacavir in HIV-positive carriers of HLA-B*57:01. peg-ifn-alpha 311-324 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 30589990-4 2019 Studies verified predisposition of HIV-positive carriers of UGT1A1*28 to severe atazanavir-induced hyperbilirubinaemia, intolerance to 5-fluorouracyl in gastrointestinal cancer patients with deleterious DPYD variants, failure of HCV-infected carriers of IFNL3 rs12979860 to obtain a sustained viral response to PEG-IFN-alpha, and hypersensitivity reactions to abacavir in HIV-positive carriers of HLA-B*57:01. abacavir 360-368 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 30810774-0 2019 Pazopanib interacts with irinotecan by inhibiting UGT1A1-mediated glucuronidation, but not OATP1B1-mediated hepatic uptake, of an active metabolite SN-38. pazopanib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 30810774-0 2019 Pazopanib interacts with irinotecan by inhibiting UGT1A1-mediated glucuronidation, but not OATP1B1-mediated hepatic uptake, of an active metabolite SN-38. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 30810774-3 2019 To clarify the possible mechanism underlying that drug-drug interaction, we investigated the potential for pazopanib to inhibit UDP-glucuronosyltransferase (UGT)1A1 and organic anion-transporting polypeptide (OATP)1B1, which are involved in detoxification and hepatic uptake of SN-38, respectively. pazopanib 107-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-164 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 253-259 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). pazopanib 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 30810774-6 2019 RESULTS: Concentration-dependent inhibition of SN-38 glucuronidation was observed in the HLMs and recombinant human UGT1A1 experiments: Pazopanib noncompetitively inhibited SN-38 glucuronidation by HLMs (Ki,u = 1.6 +- 0.05 microM) and recombinant human UGT1A1 (Ki,u = 0.69 +- 0.02 microM). pazopanib 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 253-259 30810774-10 2019 CONCLUSIONS: Results showed that pazopanib inhibits UGT1A1-mediated SN-38 glucuronidation, but not OATP1B1-mediated SN-38 uptake. pazopanib 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 30810774-10 2019 CONCLUSIONS: Results showed that pazopanib inhibits UGT1A1-mediated SN-38 glucuronidation, but not OATP1B1-mediated SN-38 uptake. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 30804050-7 2019 The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). dolutegravir 113-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 30804050-7 2019 The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). cabotegravir 155-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 30804050-7 2019 The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). cabotegravir 155-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 30804050-7 2019 The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). cabotegravir 155-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 31164525-0 2019 [FOLFIRINOX for Locally Advanced and Recurrent Pancreatic Cancer with UGT1A1 *6 and or UGT1A1*28 Polymorphisms-A Report of Two Cases]. folfirinox 1-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 30339275-7 2019 In patients receiving fluoropyrimidine/irinotecan, the incremental cost between DPYD variant and UGT1A1*28/*28 carriers and noncarriers was $2,975. 2-fluoropyrimidine 22-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 30339275-7 2019 In patients receiving fluoropyrimidine/irinotecan, the incremental cost between DPYD variant and UGT1A1*28/*28 carriers and noncarriers was $2,975. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 30339275-8 2019 This study suggests that the toxicity management costs during fluoropyrimidine-based therapy are associated with DPYD and UGT1A1*28 variants and supports the utility of genotyping. 2-fluoropyrimidine 62-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 31069991-4 2019 Hyperbilirubinemia can also be caused by impaired bilirubin conjugation due to polymorphisms and mutations in genes involved in bilirubin metabolism (eg, UGT1A1). Bilirubin 5-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 31069991-4 2019 Hyperbilirubinemia can also be caused by impaired bilirubin conjugation due to polymorphisms and mutations in genes involved in bilirubin metabolism (eg, UGT1A1). Bilirubin 50-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 30583998-8 2019 Patients treated with aspirin were significantly more common in the UGT1A1*28/*28 homozygous group than in the other groups (7/23 (30.4%) compared to 22/269 (8.2%), p = 0.001). Aspirin 22-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 30583998-9 2019 CONCLUSION: Dinucleotide polymorphism in the promoter region of the UGT1A1 gene is not associated with a specific colonic phenotype in patients with sporadic colorectal cancer. Dinucleoside Phosphates 12-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 31164525-0 2019 [FOLFIRINOX for Locally Advanced and Recurrent Pancreatic Cancer with UGT1A1 *6 and or UGT1A1*28 Polymorphisms-A Report of Two Cases]. folfirinox 1-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. Irinotecan 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 30594625-0 2019 Inhibition of UGT1A1 by natural and synthetic flavonoids. Flavonoids 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 31164525-2 2019 Since both patients were revealed to harbor UGT1A1 polymorphisms, which were highly associated with irinotecan-induced toxicity(the former: UGT1A1 *6/*28, the latter: UGT1A1*6/*6), there was no alternative hopeful treatment other than FOLFIRINOX for them. folfirinox 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 30594625-2 2019 Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. Flavonoids 45-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-122 30594625-5 2019 Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. kaempferol 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 31164525-6 2019 Since the determined regimen of FOLFIRINOX for patients with high-risk UGT1A1 polymorphisms has not been developed yet, it would be critical to accumulate and review an experience of FOLFIRINOX administration for these patients. folfirinox 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 30594625-6 2019 Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. Flavonoids 5-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 30594625-2 2019 Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. Flavonoids 45-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 30594625-2 2019 Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. Bilirubin 204-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-122 31164525-6 2019 Since the determined regimen of FOLFIRINOX for patients with high-risk UGT1A1 polymorphisms has not been developed yet, it would be critical to accumulate and review an experience of FOLFIRINOX administration for these patients. folfirinox 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 30594625-2 2019 Recent studies demonstrate that some natural flavonoids are potent inhibitors of the human UDP-glucuronosyltransferase 1A1 (UGT1A1), a key enzyme in detoxification of endogenous harmful compounds such as bilirubin. Bilirubin 204-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 30594625-3 2019 In this study, the inhibitory effects of 56 natural and synthetic flavonoids on UGT1A1 were assayed, while the structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated. Flavonoids 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 30594625-4 2019 The results demonstrated that the C-3 and C-7 hydroxyl groups on the flavone skeleton would enhance UGT1A1 inhibition, while flavonoid glycosides displayed weaker inhibitory effects than their corresponding aglycones. flavone 69-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 30594625-5 2019 Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. Flavonoids 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 30594625-5 2019 Further investigation on inhibition kinetics of two strong flavonoid-type UGT1A1 inhibitors, acacetin and kaempferol, yielded interesting results. acacetin 93-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 30594625-6 2019 Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. Flavonoids 5-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 29543105-7 2019 UGT1A6 and UGT1A1 played a major role in R/S-6- and 7-hydroxywarfarin glucuronidation, respectively, whereas UGT1A9 accounted for almost all of the generation of the R/S-8-hydroxywarfarin glucuronide. r 41-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 30594625-6 2019 Both flavonoids were competitive inhibitors against UGT1A1-mediated NHPN-O-glucuronidation, but were mixed and competitive inhibitors toward UGT1A1-mediated NCHN-O-glucuronidation, respectively. nchn-o 157-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 30594625-7 2019 Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. kaempferol 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 30594625-7 2019 Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. acacetin 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 30594625-7 2019 Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. acacetin 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 30594625-7 2019 Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. Bilirubin 172-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 30594625-7 2019 Furthermore, docking simulations showed that the binding areas of NHPN, kaempferol and acacetin on UGT1A1 were highly overlapping, and convergence with the binding area of bilirubin within UGT1A1. Bilirubin 172-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30594625-8 2019 In summary, detailed structure-inhibition relationships of flavonoids as UGT1A1 inhibitors were investigated carefully and the findings shed new light on the interactions between flavonoids and UGT1A1, and will contribute considerably to the development of flavonoid-type drugs without strong UGT1A1 inhibition. Flavonoids 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 30736072-8 2019 Glucuronidation of isoscopoletin and scopoletin was catalyzed by the human UGT1A1, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, and UGT2B17. isoscopoletin 19-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 30736072-8 2019 Glucuronidation of isoscopoletin and scopoletin was catalyzed by the human UGT1A1, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, and UGT2B17. Scopoletin 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 29543105-7 2019 UGT1A6 and UGT1A1 played a major role in R/S-6- and 7-hydroxywarfarin glucuronidation, respectively, whereas UGT1A9 accounted for almost all of the generation of the R/S-8-hydroxywarfarin glucuronide. s-6- and 7-hydroxywarfarin 43-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 30919115-7 2019 The median dose of lenvatinib in patients with UGT1A1*6/*6 or *6/*28 (poor metabolizers [PMs]) was significantly lower than that in patients with UGT1A1*1/*1 (10 and 14 mg, respectively), whereas the median bilirubin levels were significant higher in UGT1A1 PMs (0.9 and 0.5 mg/dL, respectively). lenvatinib 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 30328531-0 2019 Influence of UGT1A1 polymorphism on etoposide plus platinum-induced neutropenia in Japanese patients with small-cell lung cancer. Etoposide 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 30328531-9 2019 CONCLUSIONS: UGT1A1*28 and UGT1A1*6 mutations might be regarded as predictors for etoposide-induced grade 4 neutropenia. Etoposide 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 30328531-0 2019 Influence of UGT1A1 polymorphism on etoposide plus platinum-induced neutropenia in Japanese patients with small-cell lung cancer. Platinum 51-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 30328531-1 2019 BACKGROUND: The association between UGT1A1 polymorphism and etoposide-induced toxicities is still not clear. Etoposide 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 30328531-2 2019 The aim of this study was to assess the association between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphism and severe hematologic toxicities in Japanese patients receiving etoposide plus platinum chemotherapy for small-cell lung cancer. Etoposide 200-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-107 30328531-2 2019 The aim of this study was to assess the association between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphism and severe hematologic toxicities in Japanese patients receiving etoposide plus platinum chemotherapy for small-cell lung cancer. Etoposide 200-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 30328531-2 2019 The aim of this study was to assess the association between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphism and severe hematologic toxicities in Japanese patients receiving etoposide plus platinum chemotherapy for small-cell lung cancer. Platinum 215-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-107 30328531-2 2019 The aim of this study was to assess the association between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphism and severe hematologic toxicities in Japanese patients receiving etoposide plus platinum chemotherapy for small-cell lung cancer. Platinum 215-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 30328531-6 2019 The incidence of grade 4 neutropenia during the first cycle of etoposide-based chemotherapy was higher in patients with homozygous (hmz) polymorphisms for UGT1A1*28 and *6 (*28/*28, *6/*6, and *6/*28) than in patients with wild-type (wt) (*1/*1) and heterozygous (htz) (*1/*28 and *1/*6) polymorphisms (88% vs 43% P = 0.03). Etoposide 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 30328531-9 2019 CONCLUSIONS: UGT1A1*28 and UGT1A1*6 mutations might be regarded as predictors for etoposide-induced grade 4 neutropenia. Etoposide 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 30820183-7 2019 Pretreatment levels of ferritin and bilirubin were associated with the presence of UGT1A1*28 genotype, indicating its potential as a predictive marker. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 30628534-0 2019 Cost-effectiveness analysis of UGT1A1*6/*28 genotyping for preventing FOLFIRI-induced severe neutropenia in Chinese colorectal cancer patients. FOLFIRI regimen 70-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 29436891-8 2019 Glucuronidation of alpinetin was significantly correlated with glucuronidation of estradiol (an activity marker of UGT1A1), chenodeoxycholic acid (an activity marker of UGT1A3), propofol (an activity marker of UGT1A9) and 5-hydroxyrofecoxib (an activity marker of UGT2B15), confirming the important roles of UGT1A1, UGT1A3, UGT1A9 and UGT2B15 in alpinetin glucuronidation. alpinetin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 30466000-6 2019 Moreover, RA weakly inhibited CYP2C9 and 2E1 activities with IC50 values of 39.6 and 61.0 muM, respectively, while moderately inhibiting UGT1A1, 1A6, and 2B7 with IC50 values of 9.24, 19.1, and 23.4 muM, respectively. rosmarinic acid 10-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 29436891-8 2019 Glucuronidation of alpinetin was significantly correlated with glucuronidation of estradiol (an activity marker of UGT1A1), chenodeoxycholic acid (an activity marker of UGT1A3), propofol (an activity marker of UGT1A9) and 5-hydroxyrofecoxib (an activity marker of UGT2B15), confirming the important roles of UGT1A1, UGT1A3, UGT1A9 and UGT2B15 in alpinetin glucuronidation. alpinetin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 308-314 29436891-8 2019 Glucuronidation of alpinetin was significantly correlated with glucuronidation of estradiol (an activity marker of UGT1A1), chenodeoxycholic acid (an activity marker of UGT1A3), propofol (an activity marker of UGT1A9) and 5-hydroxyrofecoxib (an activity marker of UGT2B15), confirming the important roles of UGT1A1, UGT1A3, UGT1A9 and UGT2B15 in alpinetin glucuronidation. Estradiol 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 31633039-3 2019 Pharmacogenetics is increasingly used to stratify patients for precision medicine treatments, for instance, the UGT1A1*28 polymorphism as a dosage indicator for the use of irinotecan as well as epidermal growth factor receptor (EGFR) immunohistochemistry and KRAS Proto-Oncogene (KRAS) exon 2 mutation tests for determining the likelihood of treatment response to cetuximab or panitumumab treatment in metastatic colorectal cancer (CRC). irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 30466000-2 2019 The involvement of selected enzymes (CYP1A2, CYP2C19, CYP2E1, CYP3 A4, UGT1A1, UGT1A6, and UGT2B7) in the metabolism of RA and the inhibitory effect of RA on the enzyme activity were comprehensively evaluated using human recombinant isozyme system. rosmarinic acid 120-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 30466000-7 2019 By constructing Line weaver-Burk plots, the type of inhibition exhibited by RA on CYP and UGT activities was determined as follows: CYP2C19, mixed inhibition; CYP2E1, UGT1A1, UGT1A6, and UGT2B7, competitive inhibition. rosmarinic acid 76-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 30447099-0 2019 Impact of UGT1A1 genetic polymorphism on toxicity in unresectable pancreatic cancer patients undergoing FOLFIRINOX. folfirinox 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 30447099-1 2019 Studies have indicated an association between UDP-glucuronosyltransferase-1A1 (UGT1A1) genetic polymorphisms and irinotecan-induced toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-77 30447099-1 2019 Studies have indicated an association between UDP-glucuronosyltransferase-1A1 (UGT1A1) genetic polymorphisms and irinotecan-induced toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 31685767-0 2019 Study on the Optimal Dose of Irinotecan for Patients with Heterozygous Uridine Diphosphate-Glucuronosyltransferase 1A1 (UGT1A1). irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-118 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. folfirinox 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. Oxaliplatin 157-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. Irinotecan 170-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. Fluorouracil 182-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-2 2019 We undertook this study to investigate the association between UGT1A1 genetic polymorphisms and toxicity in patients treated with the FOLFIRINOX (comprising oxaliplatin, irinotecan, fluorouracil, and leucovorin) chemotherapy regimen in the JASPAC 06 study. Leucovorin 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 30447099-7 2019 In the original FOLFIRINOX group, the incidence of diarrhea was significantly higher among patients with UGT1A1 heterozygous type than among those with UGT1A1 WT and the incidence of leukopenia and diarrhea was significantly higher among patients with UGT1A1 -/*6 than among those with UGT1A1 -/*28. folfirinox 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 30447099-7 2019 In the original FOLFIRINOX group, the incidence of diarrhea was significantly higher among patients with UGT1A1 heterozygous type than among those with UGT1A1 WT and the incidence of leukopenia and diarrhea was significantly higher among patients with UGT1A1 -/*6 than among those with UGT1A1 -/*28. folfirinox 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 30447099-7 2019 In the original FOLFIRINOX group, the incidence of diarrhea was significantly higher among patients with UGT1A1 heterozygous type than among those with UGT1A1 WT and the incidence of leukopenia and diarrhea was significantly higher among patients with UGT1A1 -/*6 than among those with UGT1A1 -/*28. folfirinox 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 30447099-7 2019 In the original FOLFIRINOX group, the incidence of diarrhea was significantly higher among patients with UGT1A1 heterozygous type than among those with UGT1A1 WT and the incidence of leukopenia and diarrhea was significantly higher among patients with UGT1A1 -/*6 than among those with UGT1A1 -/*28. folfirinox 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 30447099-10 2019 In conclusion, patients with heterozygous UGT1A1 polymorphisms treated with the original FOLFIRINOX regimen experienced severe toxicity more frequently than patients with WT UGT1A1. folfirinox 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 30439556-6 2019 Butyrate also promoted induction of CYP1, NQO1, NAT2, UGT1A1 or UGT1A4 in human colon Caco-2 cells, in a differentiation-dependent manner. Butyrates 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30439556-10 2019 In some cases (NAT2, UGT1 A1), this was linked to inhibition of histone deacetylases (HDAC), as confirmed by using HDAC inhibitor trichostatin A. trichostatin A 130-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-28 30421550-0 2019 Prolonged central apnoea after intravenous morphine administration in a 12-year-old male with a UGT1A1 loss-of-function polymorphism. Morphine 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 29357726-7 2019 TSI could be reduced to a relatively unstable hydroquinone intermediate by NAD(P)H: quinone oxidoreductase 1 (NQO1), and then immediately conjugated with glucuronic acid by a panel of UGTs, especially UGT1A9, UGT1A1 and UGT1A8. Glucuronic Acid 154-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 29357726-8 2019 Additionally, NQO1 could also reduce hydroxylated TSI to a hydroquinone intermediate, which was immediately glucuronidated by UGT1A1. hydroquinone 59-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 30439556-4 2019 In combination with BaP, butyrate potentiated induction of cytochrome P450 family 1 enzymes (CYP1A1), aldo-keto reductases (AKR1C1) or UDP-glucuronosyltransferases (UGT1A1). Butyrates 25-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 31054979-2 2019 To examine this hypothesis, we investigated the relationship between plasma bilirubin concentrations and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variations (associated with increased bilirubin concentrations) with total/CVD and cancer mortality. Bilirubin 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 31054979-2 2019 To examine this hypothesis, we investigated the relationship between plasma bilirubin concentrations and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variations (associated with increased bilirubin concentrations) with total/CVD and cancer mortality. Bilirubin 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 31054979-2 2019 To examine this hypothesis, we investigated the relationship between plasma bilirubin concentrations and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variations (associated with increased bilirubin concentrations) with total/CVD and cancer mortality. Bilirubin 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 31054979-10 2019 The UGT1A1*28 allele, a genetic marker of raised bilirubin, was also negatively associated with total/cancer mortality, although the associations were not statistically significant. Bilirubin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 30585257-1 2019 BACKGROUND: Patients harbouring the UGT1A1*28/*28 genotype are at risk of severe toxicity with the standard irinotecan dose. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 30585257-6 2019 In the experimental group, the irinotecan dose was 300 mg/m2 for UGT1A1*1/*1 and 260 mg/m2 for *1/*28 patients. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30585257-13 2019 CONCLUSIONS: Patients with the UGT1A1*1/*1 and *1/*28 genotypes can receive high doses of irinotecan to achieve a more favourable ORR without significant adverse events. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 31685767-0 2019 Study on the Optimal Dose of Irinotecan for Patients with Heterozygous Uridine Diphosphate-Glucuronosyltransferase 1A1 (UGT1A1). irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 31685767-1 2019 Uridine 5"-diphospho-glucuronosyltransferase (UGT), a metabolic enzyme of irinotecan active metabolite, has two genetic polymorphisms (UGT1A1*6 and UGT1A1*28). irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 31685767-1 2019 Uridine 5"-diphospho-glucuronosyltransferase (UGT), a metabolic enzyme of irinotecan active metabolite, has two genetic polymorphisms (UGT1A1*6 and UGT1A1*28). irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 30421550-4 2019 EVIDENCE THAT LINKS DRUG TO EVENT: Pharmacogenomic testing revealed that the patient was homozygous for the T allele at the rs887829 SNP in UGT1A1, an enzyme involved in the metabolism of morphine. Morphine 188-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 31685767-2 2019 In UGT1A1 homozygous or heterozygous patients, metabolism is delayed and the risk of developing adverse effects is increased, and therefore, dose reduction of irinotecan is considered. irinotecan 159-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 3-9 30421550-6 2019 MECHANISM: Morphine is metabolized by UDP-glucuronosyltransferase (UGT)-2B7 and UGT1A1 to form its major metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). Morphine 11-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 31685767-11 2019 For lung cancer patients with UGT1A1 heterozygote types who start irinotecan therapy, reducing the initial dose by approximately 20% might be a safer chemotherapy without decreasing the therapeutic effect. irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 30421550-6 2019 MECHANISM: Morphine is metabolized by UDP-glucuronosyltransferase (UGT)-2B7 and UGT1A1 to form its major metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). morphine-3-glucuronide 117-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 30377777-11 2019 CONCLUSION: The present study demonstrated that dose reduction by 20% ensured safety and efficacy of irinotecan in mCRC patients with homozygous mutation in UGT1A1 genes. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 30421550-6 2019 MECHANISM: Morphine is metabolized by UDP-glucuronosyltransferase (UGT)-2B7 and UGT1A1 to form its major metabolites morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). morphine-6-glucuronide 150-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 30421550-7 2019 Our patient was a poor metabolizer through UGT1A1, likely leading to increased respiratory depression as morphine has greater respiratory depressant effects compared to its metabolites. Morphine 105-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 30377777-0 2019 Dose adjustment of irinotecan based on UGT1A1 polymorphisms in patients with colorectal cancer. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 29932028-4 2019 SN-38 is detoxified by the formation of SN-38 glucuronide, through UGT1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 30377777-2 2019 SN-38 is an active metabolite of irinotecan, which is formed by carboxylesterase and inactivated by UDP-glucuronyltransferase (UGT) 1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-135 30377777-2 2019 SN-38 is an active metabolite of irinotecan, which is formed by carboxylesterase and inactivated by UDP-glucuronyltransferase (UGT) 1A1. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-135 30377777-3 2019 The UGT enzyme activity is reduced in patients with homozygous mutation in UGT1A1 genes (*6/*6, *28/*28 and *6/*28); thus dose reduction is required for prevention of severe adverse events associated with irinotecan. Irinotecan 205-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 30377777-4 2019 The present study was designed to investigate the relationship between UGT1A1 polymorphisms and the incidence of adverse events or the therapeutic effect in mCRC patients who received irinotecan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29932028-4 2019 SN-38 is detoxified by the formation of SN-38 glucuronide, through UGT1A1. 7-ethyl-10-hydroxycamptothecin glucuronide 40-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 29932028-5 2019 Genetic polymorphisms in the UGT1A1 gene are associated to higher exposures to SN-38 and severe toxicity. Irinotecan 79-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 30171692-0 2018 CYP3A4-mediated effects of rifampicin on the pharmacokinetics of vilaprisan and its UGT1A1-mediated effects on bilirubin glucuronidation in humans. Bilirubin 111-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 98-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 98-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 30619479-9 2018 The variants of UGT1A1 *28 and UGT1A1 *6 were strongly associated with increased total bilirubin (TB), direct bilirubin (DB), and indirect bilirubin (IB) levels (each P < 0.001). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30385458-4 2019 CN-1 is an autosomal recessive disorder caused by damaging mutations in the gene for UGT1A1, the hepatic enzyme responsible for bilirubin conjugation in humans. Bilirubin 128-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 30527657-1 2019 Differences in drug metabolism associated with UGT1A1 polymorphism could result in individualized local response to hepatic chemoembolization with irinotecan-eluting beads (DEBIRI) or predictable toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 30397001-7 2019 Moreover, CCAR1 could be recruited to the gtPBREM of the UGT1A1 enhancer by CAR but not to the PBREM of the CYP2B6 enhancer. gtpbrem 42-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 30599907-12 2019 CONCLUSION: The results indicated that the HF originated overexpression of Ugt1a1, Ugt1a7, and P-gp level played important roles in pharmacokinetics of puerarin, suggested the clinical regimen of puerarin based on normal populations might be inappropriate for patients with chronic liver diseases. puerarin 152-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 30599907-12 2019 CONCLUSION: The results indicated that the HF originated overexpression of Ugt1a1, Ugt1a7, and P-gp level played important roles in pharmacokinetics of puerarin, suggested the clinical regimen of puerarin based on normal populations might be inappropriate for patients with chronic liver diseases. puerarin 196-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 30237061-0 2018 The roles of breast cancer resistance protein (BCRP/ABCG2) and multidrug resistance-associated proteins (MRPs/ABCCs) in the excretion of cycloicaritin-3-O-glucoronide in UGT1A1-overexpressing HeLa cells. cycloicaritin-3-o-glucoronide 137-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 30237061-4 2018 First, beta-estradiol was used to validate the expression of active UGT1A1 protein in engineered HeLa1A1 cells. Estradiol 7-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 30237061-9 2018 Meanwhile, the UGT1A1 modified HeLa cells were a simple and practical tool to study UGT1A1-mediated glucuronidation and to characterize BCRP and MRPs-mediated glucuronide transport at a cellular level. Glucuronides 159-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 30171692-2 2018 In addition, the effects of rifampicin on the glucuronidation of bilirubin, an endogenous UDP-glucuronosyltransferase family 1 member A1 (UGT1A1) substrate, were explored. Bilirubin 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-136 30246377-0 2018 UGT1A1 polymorphisms in rectal cancer associated with the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30246377-11 2018 Our results indicate that UGT1A1 polymorphism is a predictive factor to determine the clinical efficacy of preoperative chemoradiotherapy and hematological toxicity induced by chemoradiotherapy using irinotecan in locally advanced rectal cancer patients. Irinotecan 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 30171692-2 2018 In addition, the effects of rifampicin on the glucuronidation of bilirubin, an endogenous UDP-glucuronosyltransferase family 1 member A1 (UGT1A1) substrate, were explored. Bilirubin 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 30246377-1 2018 The purpose of the present study was to assess the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan against locally advanced lower rectal cancer according to UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-213 30246377-1 2018 The purpose of the present study was to assess the efficacy and toxicity of preoperative chemoradiotherapy using irinotecan against locally advanced lower rectal cancer according to UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 215-221 30171692-10 2018 Further, it was associated with an increase in bilirubin glucuronidation, indicating that UGT1A1 was induced. Bilirubin 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 30105552-6 2018 This study demonstrates that different variations present in the UGT1A1 gene and specifically, the H55R variation had a significant effect on bilirubin levels and could be genetic risk factors for hyperbilirubinemia. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 30194276-0 2018 6-Chloro-5-[4-(1-Hydroxycyclobutyl)Phenyl]-1H-Indole-3-Carboxylic Acid is a Highly Selective Substrate for Glucuronidation by UGT1A1, Relative to beta-Estradiol. PF-6409577 0-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 30194276-0 2018 6-Chloro-5-[4-(1-Hydroxycyclobutyl)Phenyl]-1H-Indole-3-Carboxylic Acid is a Highly Selective Substrate for Glucuronidation by UGT1A1, Relative to beta-Estradiol. Estradiol 146-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 30194276-4 2018 Studies using a panel of 13 human recombinant UGT (hrUGT) enzymes indicated that PF-06409577 was converted to M1 in a highly selective fashion by UGT1A1, which was further verified in human liver microsomes treated with specific chemical inhibitors, and in different UGT1A1 expressers. pyrazofurin 81-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 30194276-4 2018 Studies using a panel of 13 human recombinant UGT (hrUGT) enzymes indicated that PF-06409577 was converted to M1 in a highly selective fashion by UGT1A1, which was further verified in human liver microsomes treated with specific chemical inhibitors, and in different UGT1A1 expressers. pyrazofurin 81-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 267-273 30194276-5 2018 Conversion of PF-06409577 to M1 by UGT1A1 occurred in a relatively selective fashion, compared with beta-estradiol (ES), a conventional probe substrate of UGT1A1. PF-6409577 14-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 30194276-5 2018 Conversion of PF-06409577 to M1 by UGT1A1 occurred in a relatively selective fashion, compared with beta-estradiol (ES), a conventional probe substrate of UGT1A1. PF-6409577 14-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 30194276-9 2018 Our results suggest the potential utility of PF-06409477 as a selective probe UGT1A1 substrate for UGT reaction phenotyping and inhibition studies in preclinical discovery/development. pf-06409477 45-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 30527181-0 2018 Association of nephrotoxicity during platinum-etoposide doublet therapy with UGT1A1 polymorphisms in small cell lung cancer patients. Platinum 37-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 30527181-0 2018 Association of nephrotoxicity during platinum-etoposide doublet therapy with UGT1A1 polymorphisms in small cell lung cancer patients. Etoposide 46-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Etoposide 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-53 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Etoposide 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Etoposide 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-53 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Etoposide 231-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-53 30527181-2 2018 Uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1) is thought to be largely responsible for the glucuronidation of etoposide as well as that of irinotecan, suggesting that polymorphisms of UGT1A1 might be predictive of etoposide toxicity. Etoposide 231-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30527181-10 2018 CONCLUSION: Our results reveal an association between UGT1A1 polymorphisms and toxicity of platinum-etoposide doublet therapy in SCLC patients, suggesting that close monitoring for toxicity, especially nephrotoxicity, is warranted for patients with such variant alleles receiving this treatment. Platinum 91-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30527181-10 2018 CONCLUSION: Our results reveal an association between UGT1A1 polymorphisms and toxicity of platinum-etoposide doublet therapy in SCLC patients, suggesting that close monitoring for toxicity, especially nephrotoxicity, is warranted for patients with such variant alleles receiving this treatment. Etoposide 100-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30563996-1 2018 Background/objectives: Individuals with Gilbert"s syndrome (GS) harbor mutations in the UGT1A1 gene and are known to have elevated levels of bilirubin, which enhances the risk for gall stone formation. Bilirubin 141-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 9-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 29955115-0 2018 UGT1A1 polymorphisms associated with prolactin response in risperidone-treated children and adolescents with autism spectrum disorder. Risperidone 59-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 29955115-8 2018 In this DMET microarray platform, we found three UGT1A1 variants with suggestive evidences of association with the risperidone-induced prolactin response both measured by hyperprolactinemia and by prolactin level. Risperidone 115-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 30538568-0 2018 Value of plasma SN-38 levels and DPD activity in irinotecan-based individualized chemotherapy for advanced colorectal cancer with heterozygous type UGT1A1*6 or UGT1A1*28. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 30538568-0 2018 Value of plasma SN-38 levels and DPD activity in irinotecan-based individualized chemotherapy for advanced colorectal cancer with heterozygous type UGT1A1*6 or UGT1A1*28. Irinotecan 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 9-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 9-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 9-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-3 2018 Results: CSN-38 1.5h and CSN-38 49h of the UGT1A1*6 or UGT1A1*28 heterozygous type were close to those of UGT1A1*6 and UGT1A1*28 wild-types; some of those with relatively high CSN-38 1.5h levels obtained better median progression-free survival (mPFS), whereas others with higher CSN-38 49h concentrations showed a relatively high incidence of adverse reactions possibly because of the decreased activity of DPD. 4-nitrocatechol sulfate 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 30538568-5 2018 For cases with comparably low DPD activity, advisable primary and subsequent dose adjustment of 5-fluorouracil based on plasma 5-fluorouracil levels may be a practical strategy for reducing the occurrence of adverse reactions for personalized treatment of the UGT1A1*6 or UGT1A1*28 heterozygous type. Fluorouracil 96-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 260-266 30538568-5 2018 For cases with comparably low DPD activity, advisable primary and subsequent dose adjustment of 5-fluorouracil based on plasma 5-fluorouracil levels may be a practical strategy for reducing the occurrence of adverse reactions for personalized treatment of the UGT1A1*6 or UGT1A1*28 heterozygous type. Fluorouracil 96-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 272-278 30538568-5 2018 For cases with comparably low DPD activity, advisable primary and subsequent dose adjustment of 5-fluorouracil based on plasma 5-fluorouracil levels may be a practical strategy for reducing the occurrence of adverse reactions for personalized treatment of the UGT1A1*6 or UGT1A1*28 heterozygous type. Fluorouracil 127-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 260-266 30538568-5 2018 For cases with comparably low DPD activity, advisable primary and subsequent dose adjustment of 5-fluorouracil based on plasma 5-fluorouracil levels may be a practical strategy for reducing the occurrence of adverse reactions for personalized treatment of the UGT1A1*6 or UGT1A1*28 heterozygous type. Fluorouracil 127-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 272-278 32186076-6 2018 YMJHT inhibited CYP2E1 activity, with a negligible inhibition on the activities of CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4, UGT1A1, UGT1A4 and UGT2B7. ymjht 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 29932297-0 2018 UGT1A1*6 and UGT1A1*28 polymorphisms are correlated with irinotecan-induced toxicity: A meta-analysis. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 29932297-0 2018 UGT1A1*6 and UGT1A1*28 polymorphisms are correlated with irinotecan-induced toxicity: A meta-analysis. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 29932297-1 2018 BACKGROUND: Previous articles explored the role of UGT1A1 polymorphism on predicting irinotecan-induced toxicity, but the conclusions were still inconsistent and not comprehensive. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 29932297-2 2018 We performed this meta-analysis to investigate the association between UGT1A1 polymorphism and irinotecan-induced toxicity. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29932297-8 2018 Both UGT1A1*6 and UGT1A1*28 polymorphism are significantly associated with severe irinotecan-induced toxicity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 29932297-8 2018 Both UGT1A1*6 and UGT1A1*28 polymorphism are significantly associated with severe irinotecan-induced toxicity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 29932297-12 2018 Subgroup analysis exhibited that for UGT1A1*6 polymorphism, patients treated with low-dose irinotecan were at a notable risk of toxicity. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 29932297-13 2018 Moreover, the association between UGT1A1*6 polymorphism and irinotecan-induced toxicity was found in patients suffering from respiratory system cancers. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29932297-14 2018 CONCLUSIONS: Both UGT1A1*6 and UGT1A1*28 polymorphisms can be considered as predictors of irinotecan-induced toxicity, with effect varying by race, cancer type and irinotecan dose. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 29932297-14 2018 CONCLUSIONS: Both UGT1A1*6 and UGT1A1*28 polymorphisms can be considered as predictors of irinotecan-induced toxicity, with effect varying by race, cancer type and irinotecan dose. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 30266131-1 2018 Crigler-Najjar syndrome type II is caused by mutations in the UGT1A1 gene resulting in severely reduced hepatic activity of UDP-glucoronyltransferase - an enzyme required to convert bilirubin into a more soluble form that can then be removed from the body. Bilirubin 182-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 29309376-8 2018 In addition, we analyzed bilirubin uridine diphosphate (UDP)-glucuronosyltransferase 1A1 gene that revealed a heterozygous state ([TA]6/[TA]7). Bilirubin 25-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-88 29273261-1 2018 PURPOSE: We aimed to identify the maximum tolerated dose (MTD) of weekly irinotecan in combination with capecitabine-based neoadjuvant chemoradiation according to the UGT1A1*28 genotype in patients with locally advanced rectal cancer. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 29273261-11 2018 CONCLUSION: A higher dose of weekly irinotecan in combination with capecitabine-based CRT is feasible under the guidance of the UGT1A1*28 genotype. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 29273261-11 2018 CONCLUSION: A higher dose of weekly irinotecan in combination with capecitabine-based CRT is feasible under the guidance of the UGT1A1*28 genotype. Capecitabine 67-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 30036684-4 2018 Herein, we designed and optimized a validated cocktail method for the simultaneous evaluation of drug-mediated inhibition of the main five UGT isoforms using respective specific probe substrates (estradiol for UGT1A1, chenodeoxycholic acid for UGT1A3, serotonin for UGT1A6, propofol for UGT1A9/PROG and zidovudine for UGT2B7/AZTG) in human and rat liver microsomes by liquid chromatography-tandem mass spectrometry (LCMS/MS). Estradiol 196-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 210-216 29951694-1 2018 PURPOSE: Belinostat is a second-generation histone deacetylase inhibitor (HDI) predominantly metabolized by UGT1A1-mediated glucuronidation. belinostat 9-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 30139029-0 2018 [Relationship between UGT1A1 gene polymorphisms and irinotecan-induced severe adverse events]. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 30139029-1 2018 Objective: To investigate the relationship between UGT1A1*6, UGT1A1*28, UGT1A1*60 and UGT1A1*93 polymorphisms and irinotecan-induced severe adverse reactions(grade 3-4 delayed diarrhea and neutropenia) in Chinese cancer patients. Irinotecan 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 30139029-16 2018 Conclusion: The cancer patients who carried UGT1A1*6, UGT1A1*28 and UGT1A1*60 gene polymorphisms have high risk of severe adverse events caused by irinotecan-based chemotherapy. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 29735754-5 2018 Conversely, PT-GP and PT-Ac caused direct inhibition of almost all P450 and UGT enzymes, with CYP1A2 (IC50 values of 0.8-4.2 muM), CYP2C8 (IC50 values of 1.1-12 muM), and UGT1A1 (IC50 values of 4.5-5.4 muM) inhibited to the greatest extent. pt-gp 12-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 29735754-5 2018 Conversely, PT-GP and PT-Ac caused direct inhibition of almost all P450 and UGT enzymes, with CYP1A2 (IC50 values of 0.8-4.2 muM), CYP2C8 (IC50 values of 1.1-12 muM), and UGT1A1 (IC50 values of 4.5-5.4 muM) inhibited to the greatest extent. 6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo(3.2.1)octane 22-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 29909091-2 2018 We assessed potential clinical variables that may predict toxicity and more specifically the role of UGT1A1 polymorphisms associated with irinotecan toxicity. Irinotecan 138-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-109 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Fluorouracil 198-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-109 30498448-4 2018 Genetic variants resulting in decreased enzymatic activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and dihydropyrimidine dehydrogenase (DPD) are known to increase irinotecan and 5-fluorouracil-related toxicity, respectively. Fluorouracil 198-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 30334318-0 2018 Efflux excretion of bisdemethoxycurcumin-O-glucuronide in UGT1A1-overexpressing HeLa cells: Identification of breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins 1 (MRP1) as the glucuronide transporters. bisdemethoxycurcumin-o-glucuronide 20-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 30105461-2 2018 Therefore, it would be beneficial to understand whether there is a relationship between inhibition of uridine-5"-diphosphate glucuronosyltransferase (UGT) 1A1 activity and observed bilirubin elevations in TKI drug-treated patients. Bilirubin 181-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-158 30105461-3 2018 UGT1A1 is responsible for the glucuronidation of bilirubin which leads to its elimination in the bile. Bilirubin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 30425912-1 2018 The UGT1A1 enzyme is involved in the metabolism of bilirubin and numerous medications. Bilirubin 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 30425912-11 2018 Level of bilirubin in pediatric GS patients at diagnosis was UGT1A1 (TA)n promoter genotype-dependent. Bilirubin 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 30151596-7 2018 Moreover, we demonstrate that UGT1A1, 1A6 and 1A9 proteins catalyze the synthesis of CHZ-O-Glc while CHZ-N-Glc is produced by UGT1A9 specifically. chz-o-glc 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 29934866-5 2018 The C-terminal domain is identical for all UGT1A family members and required for binding to UDP-glucuronic acid as well as involved in contacts with substrates. Uridine Diphosphate Glucuronic Acid 92-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-48 30075316-3 2018 Of the 10 tested UGT isoforms, uva-ursi extracts exerted the strongest inhibitory effect on UGT1A1-mediated beta-estradiol 3-glucuronidation with the lowest IC50 value of 8.45 +- 1.56 mug/mL. Estradiol 108-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 30075316-7 2018 Thus, in vitro UGT1A1 inhibitory potentials of uva-ursi extracts might primarily result from the inhibitory activities of gallotannin and 1,2,3,6-tetragalloylglucose present in the extracts. Hydrolyzable Tannins 122-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 30075316-7 2018 Thus, in vitro UGT1A1 inhibitory potentials of uva-ursi extracts might primarily result from the inhibitory activities of gallotannin and 1,2,3,6-tetragalloylglucose present in the extracts. 1,2,3,6-tetragalloylglucose 138-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 30117422-1 2018 The adverse reaction to irinotecan is related to the single nucleotide polymorphism (SNP) of UGT1A1*6 genotype. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 30016963-0 2018 Influence of UGT1A1 polymorphisms on the outcome of acute myeloid leukemia patients treated with cytarabine-base regimens. Cytarabine 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 28891378-15 2018 Ritonavir and cobicistat are unlikely to produce clinically important drug interactions involving drugs metabolized to glucuronide conjugates by UGT1A1, 1A3, 1A6, 1A9, 2B4 and 2B7. Glucuronides 119-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 30016963-1 2018 BACKGROUNDS: UDP-glucuronosyltransferase 1A subfamily (UGT1A) enzymes can inactivate cytarabine (Ara-C) by glucuronidation, and thus serves as candidate genes for interindividual difference in Ara-C response. Cytarabine 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-43 30016963-1 2018 BACKGROUNDS: UDP-glucuronosyltransferase 1A subfamily (UGT1A) enzymes can inactivate cytarabine (Ara-C) by glucuronidation, and thus serves as candidate genes for interindividual difference in Ara-C response. Cytarabine 97-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-43 30016963-1 2018 BACKGROUNDS: UDP-glucuronosyltransferase 1A subfamily (UGT1A) enzymes can inactivate cytarabine (Ara-C) by glucuronidation, and thus serves as candidate genes for interindividual difference in Ara-C response. Cytarabine 193-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-43 30016963-3 2018 METHODS: UGT1A1*6 and *28 polymorphisms resulting in reduced UGT1A1 activity were genotyped in 726 adult acute myeloid leukemia (AML) patients treated with Ara-C based regimens. Cytarabine 156-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 30016963-5 2018 RESULTS: After one or two courses of Ara-C based induction chemotherapy, the complete remission (CR) rate was significantly higher in patients carrying the UGT1A1*6 (77.0%) or the UGT1A1*28 (76.4%) alleles as compared with corresponding wild-type homozygotes (66.9 and 68.5%, respectively). Cytarabine 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 30016963-5 2018 RESULTS: After one or two courses of Ara-C based induction chemotherapy, the complete remission (CR) rate was significantly higher in patients carrying the UGT1A1*6 (77.0%) or the UGT1A1*28 (76.4%) alleles as compared with corresponding wild-type homozygotes (66.9 and 68.5%, respectively). Cytarabine 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 30016963-7 2018 CONCLUSION: Our results suggest that UGT1A1*28 and UGT1A1*6 are associated with improved clinical outcomes in Chinese AML patients treated with Ara-C. Cytarabine 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 30016963-7 2018 CONCLUSION: Our results suggest that UGT1A1*28 and UGT1A1*6 are associated with improved clinical outcomes in Chinese AML patients treated with Ara-C. Cytarabine 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 29504153-0 2018 Relevance of CYP3A4*20, UGT1A1*37 and UGT1A1*28 variants in irinotecan-induced severe toxicity. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 30075835-8 2018 Pharmacogenomics guidelines have been published by some international scientific consortia such as the Clinical Pharmacogenomics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG) strongly suggesting a pre-treatment dose adjustment of irinotecan based on UGT1A1*28 genotype and of fluoropyrimidines based on some DPYD genetic variants, to increase treatment safety. Irinotecan 269-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 289-295 29584932-5 2018 The structure-dependent effects of the hydroxyl flavonoids on induction of UGT1A1 were similar to that observed for induction of CYP1A1 except that luteolin and apigenin induced UGT1A1 levels similar to that observed for TCDD, whereas both compounds were AhR antagonists for CYP1A1. Flavonoids 48-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 29847509-9 2018 As a positive control, baseline plasma bilirubin concentration was associated with predicted liver UGT1A1 expression level (P=1.9x10(-27)). Bilirubin 39-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 29420340-11 2018 Patients carrying more than three copies of variant UGT1A1 (*28 and *60) had higher serum levels of belinostat because of slower clearance. belinostat 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 29504153-0 2018 Relevance of CYP3A4*20, UGT1A1*37 and UGT1A1*28 variants in irinotecan-induced severe toxicity. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 29504153-1 2018 Severe irinotecan-induced toxicity is associated with UGT1A1 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 29094205-0 2018 Association between alanine aminotransferase elevation and UGT1A1*6 polymorphisms in daclatasvir and asunaprevir combination therapy for chronic hepatitis C. BACKGROUND: Liver damage presented as alanine aminotransferase (ALT) elevation and high ALT-caused treatment discontinuation occurs with high frequency in Japanese patients receiving daclatasvir plus asunaprevir (DCV/ASV) therapy for hepatitis C virus (HCV) infection, and its mechanism is unknown. daclatasvir 85-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 29637732-2 2018 Delivery of hUGT1A1-modRNA (a modified messenger RNA encoding for UGT1A1) as a lipid nanoparticle is anticipated to restore hepatic expression of UGT1A1, allowing normal glucuronidation and clearance of bilirubin in patients. Bilirubin 203-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-19 29637732-2 2018 Delivery of hUGT1A1-modRNA (a modified messenger RNA encoding for UGT1A1) as a lipid nanoparticle is anticipated to restore hepatic expression of UGT1A1, allowing normal glucuronidation and clearance of bilirubin in patients. Bilirubin 203-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 29637732-2 2018 Delivery of hUGT1A1-modRNA (a modified messenger RNA encoding for UGT1A1) as a lipid nanoparticle is anticipated to restore hepatic expression of UGT1A1, allowing normal glucuronidation and clearance of bilirubin in patients. Bilirubin 203-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 29094205-0 2018 Association between alanine aminotransferase elevation and UGT1A1*6 polymorphisms in daclatasvir and asunaprevir combination therapy for chronic hepatitis C. BACKGROUND: Liver damage presented as alanine aminotransferase (ALT) elevation and high ALT-caused treatment discontinuation occurs with high frequency in Japanese patients receiving daclatasvir plus asunaprevir (DCV/ASV) therapy for hepatitis C virus (HCV) infection, and its mechanism is unknown. asunaprevir 101-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. daclatasvir 200-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. daclatasvir 200-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. asunaprevir 204-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. asunaprevir 204-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. daclatasvir 103-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. asunaprevir 107-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. daclatasvir 122-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. asunaprevir 126-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. 8-cyclohexyl-N-((dimethylamino)sulfonyl)-1,1a,2,12b-tetrahydro-11-methoxy-1a-((3-methyl-3,8-diazabicyclo(3.2.1)oct-8-yl)carbonyl)cycloprop(d)indolo(2,1-a)(2)benzazepine-5-carboxamide 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29743555-1 2018 The aim of this study was to evaluate the effect of UGT1A1 polymorphisms on Raltegravir (RAL) and its metabolite RAL-glucuronide trough plasma concentrations ([RAL]plasma and [RAL-glu]plasma) and on the metabolic ratio (MR): [RAL-glu]plasma/[RAL]plasma. Glutamic Acid 117-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 29587224-6 2018 Besides, icaritin glucuronidation is mediated by multiple enzymes, with UGT1A1 the principal metabolizing enzyme (total CLint,UGT1A1 = 6.38 mL/mg/min). icaritin 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 29587224-6 2018 Besides, icaritin glucuronidation is mediated by multiple enzymes, with UGT1A1 the principal metabolizing enzyme (total CLint,UGT1A1 = 6.38 mL/mg/min). icaritin 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 29743555-0 2018 Impact of UGT1A1 polymorphisms on Raltegravir and its glucuronide plasma concentrations in a cohort of HIV-1 infected patients. Raltegravir Potassium 34-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 28548030-5 2018 The results showed that 100 muM of everolimus exerted more than 80% inhibition toward UGT1A1, UGT-1A3 and UGT-2B7. Everolimus 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 29743555-0 2018 Impact of UGT1A1 polymorphisms on Raltegravir and its glucuronide plasma concentrations in a cohort of HIV-1 infected patients. Glucuronides 54-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 29743555-1 2018 The aim of this study was to evaluate the effect of UGT1A1 polymorphisms on Raltegravir (RAL) and its metabolite RAL-glucuronide trough plasma concentrations ([RAL]plasma and [RAL-glu]plasma) and on the metabolic ratio (MR): [RAL-glu]plasma/[RAL]plasma. Raltegravir Potassium 76-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 29743555-1 2018 The aim of this study was to evaluate the effect of UGT1A1 polymorphisms on Raltegravir (RAL) and its metabolite RAL-glucuronide trough plasma concentrations ([RAL]plasma and [RAL-glu]plasma) and on the metabolic ratio (MR): [RAL-glu]plasma/[RAL]plasma. ral-glucuronide 113-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 28548030-8 2018 The inhibition kinetic parameters (Ki) were calculated to be 2.3, 0.07 and 4.4 muM for the inhibition of everolimus toward UGT1A1, UGT-1A3 and UGT-2B7, respectively. Everolimus 105-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 28520360-0 2012 Irinotecan Therapy and UGT1A1 Genotype Irinotecan is a topoisomerase inhibitor that is widely used in the treatment of cancer. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 29702735-2 2018 We investigated whether the variant genotypes of metabolic enzymes UGT1A1, CYP3A5 and transporter ABCB1 influence pharmacokinetic phenotype of substrate mizolastine in Chinese volunteers. mizolastine 153-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 28520360-3 2012 Irinotecan is metabolized and inactivated by an UDP-glucuronosyltransferase enzyme encoded by the gene UGT1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 28520360-5 2012 Variants of this gene, such as UGT1A1*28, are associated with reduced enzyme activity and an increased risk of irinotecan toxicity. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 28520360-6 2012 Approximately 10% of North Americans are homozygous for the UGT1A1*28 allele and are more likely to develop neutropenia following irinotecan therapy (3). Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 28520360-7 2012 The FDA-approved drug label for irinotecan states that "when administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection should be considered for patients known to be homozygous for the UGT1A1*28 allele. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 28520360-7 2012 The FDA-approved drug label for irinotecan states that "when administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection should be considered for patients known to be homozygous for the UGT1A1*28 allele. Irinotecan 151-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 28520360-9 2012 A guideline from the Dutch Pharmacogenetics Working Group (KNMP) mentions "although results are not consistent, there is sufficient evidence that a reduction in the initial dose by 30% is required for regimens containing >250 mg/m(2) of irinotecan prescribed to homozygous carriers of the UGT1A1*28 allele. Irinotecan 240-250 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 292-298 29328989-4 2018 100 muM phthalate monoesters exhibited negligible inhibition towards the activity of UGT1A1, UGT1A3, UGT1A6, UGT1A8, UGT1A10, UGT2B4, UGT2B7, UGT2B15 and UGT2B17. phthalic acid 8-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 29161598-10 2018 However, selenium supplementation led to elevations of selenium, Dio1 and TTR, and reductions of Ugt1a1, Sult1e1, CYP2b1, and TRHr. Selenium 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 29311138-0 2018 Bacterial Outer Membrane Vesicles from Dextran Sulfate Sodium-Induced Colitis Differentially Regulate Intestinal UDP-Glucuronosyltransferase 1A1 Partially Through Toll-Like Receptor 4/Mitogen-Activated Protein Kinase/Phosphatidylinositol 3-Kinase Pathway. Dextran Sulfate 39-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-144 29220881-6 2018 RESULTS: As expected, UGT1A1 genotypes were associated with baseline bilirubin levels (*1/*1 genotype: 9.1+-4.6 micromol/L; *1/*28 genotype: 10.8+-5.3; *28/*28: 16.9+-9.2; p<0.001). Bilirubin 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 29220881-8 2018 In a multivariate regression analysis adjusting for age, sex, smoking, type 2 diabetes, dyslipidemia, alanine aminotransferase (ALT) levels and bilirubin levels, the UGT1A1*28 variant predicted lower overall mortality (hazard ratio [HR], 0.86; 95% confidence interval [CI], 0.78-0.95; p=0.003). Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 29220881-11 2018 The protective effect of the UGT1A1*28 variant likely includes mechanism other than bilirubin metabolism. Bilirubin 84-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 29524031-0 2018 Contribution of UGT1A1 genetic polymorphisms related to axitinib pharmacokinetics to safety and efficacy in patients with renal cell carcinoma. Axitinib 56-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 29524031-5 2018 Genetic polymorphisms related to axitinib pharmacokinetics, including SLCO1B1, SLCO1B3, SLCO2B1, ABCB1, ABCG2, CYP2C19, CYP3A5, and UGT1A1, were analyzed. Axitinib 33-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 29524031-9 2018 Genetic polymorphisms in UGT1A1 were significantly associated with the plasma axitinib level. Axitinib 78-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 29318243-0 2018 Chemical inhibition and stable knock-down of efflux transporters leads to reduced glucuronidation of wushanicaritin in UGT1A1-overexpressing HeLa cells: the role of breast cancer resistance protein (BCRP) and multidrug resistance-associated proteins (MRPs) in the excretion of glucuronides. wushanicaritin 101-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 29223776-0 2018 Impact of edaphic factors and nutrient management on the hepatoprotective efficiency of Carlinoside purified from pigeon pea leaves: An evaluation of UGT1A1 activity in hepatitis induced organelles. carlinoside 88-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 28285550-13 2018 Stronger hydrogen bonds and hydrophobic interaction between T4 and activity cavity of UGT1A1 than T3 contributed to stronger inhibition of T4 towards UGT1A1. Hydrogen 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 28285550-13 2018 Stronger hydrogen bonds and hydrophobic interaction between T4 and activity cavity of UGT1A1 than T3 contributed to stronger inhibition of T4 towards UGT1A1. Hydrogen 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 29425147-5 2018 In human liver microsomes (HLMs), sauchinone inhibited activities of UGT1A1, 1A3, 1A6, and 2B7 with IC50 values of 8.83, 43.9, 0.758, and 0.279 muM, respectively. sauchinone 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 29223776-7 2018 This significantly up-regulated the carlinoside induced expression of the bilirubin-solubilizing UGT1A1enzyme in HepG2 cell and rat liver. carlinoside 36-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 29223776-7 2018 This significantly up-regulated the carlinoside induced expression of the bilirubin-solubilizing UGT1A1enzyme in HepG2 cell and rat liver. Bilirubin 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 29114966-5 2018 The UGT1A1 (TA)6 /(TA)8 heterozygote showed statistically significant associations with HbA2 levels (P = .019), HbF percentage (P < .001), haemoglobin levels (P = .008), PCV values (P = .007) and RBC counts (P = .041). penciclovir 173-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-57 29239247-7 2018 The rates of 7TA carriers of UGT1A1 rs8175347 in all three groups were significantly higher than the other genotypes. SCHEMBL3705488 13-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 29124328-1 2018 PURPOSE: We aimed to retrospectively evaluate the efficacy and toxicity of an irinotecan hydrochloride (CPT) and nedaplatin (N) combination therapy for recurrent and refractory endometrial carcinoma, administered based on UGT1A1 genotype. Irinotecan 78-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 29124328-1 2018 PURPOSE: We aimed to retrospectively evaluate the efficacy and toxicity of an irinotecan hydrochloride (CPT) and nedaplatin (N) combination therapy for recurrent and refractory endometrial carcinoma, administered based on UGT1A1 genotype. cpt 104-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 29124328-9 2018 Patients with a wild-type UGT1A1 status received higher doses of CPT-11 (p = 0.048) and had similar RR and CBR compared to those with a UGT1A1*6 and *28 status. Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 29589315-7 2018 It has been established that UGT1A1*28 polymorphism is associated with irinotecan toxicity, but this test is rarely performed as the management strategy has not been identified. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 29117017-9 2018 CONCLUSION: Among patients who initiated atazanavir/ritonavir-containing regimens, UGT1A1 slow metabolizer genotype rs887829 T/T was associated with increased bilirubin-related discontinuation of atazanavir in White but not in Black patients, this despite T/T genotype being more frequent in Black patients. Atazanavir Sulfate 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 29117017-9 2018 CONCLUSION: Among patients who initiated atazanavir/ritonavir-containing regimens, UGT1A1 slow metabolizer genotype rs887829 T/T was associated with increased bilirubin-related discontinuation of atazanavir in White but not in Black patients, this despite T/T genotype being more frequent in Black patients. Ritonavir 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 29117017-9 2018 CONCLUSION: Among patients who initiated atazanavir/ritonavir-containing regimens, UGT1A1 slow metabolizer genotype rs887829 T/T was associated with increased bilirubin-related discontinuation of atazanavir in White but not in Black patients, this despite T/T genotype being more frequent in Black patients. Bilirubin 159-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 29117017-9 2018 CONCLUSION: Among patients who initiated atazanavir/ritonavir-containing regimens, UGT1A1 slow metabolizer genotype rs887829 T/T was associated with increased bilirubin-related discontinuation of atazanavir in White but not in Black patients, this despite T/T genotype being more frequent in Black patients. Atazanavir Sulfate 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 29052349-0 2018 Relationship between UGT1A1*27 and UGT1A1*7 polymorphisms and irinotecan-related toxicities in patients with lung cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 29052349-0 2018 Relationship between UGT1A1*27 and UGT1A1*7 polymorphisms and irinotecan-related toxicities in patients with lung cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 29052349-1 2018 BACKGROUND: The objective of this study was to evaluate the effects of gene polymorphisms, including UGT1A1*7, *27, and *29, on the safety of irinotecan therapy. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 29052349-8 2018 SN-38 tended to remain in the blood for a prolonged period after the infusion of irinotecan in patients with UGT1A1*27 or UGT1A1*28 polymorphisms. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 29052349-10 2018 CONCLUSION: UGT1A1*27 can occur separately from UGT1A1*28 and is related to leukopenia during irinotecan treatment. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 29052349-10 2018 CONCLUSION: UGT1A1*27 can occur separately from UGT1A1*28 and is related to leukopenia during irinotecan treatment. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29052349-11 2018 UGT1A1*7 is less relevant to irinotecan-induced toxicities, and UGT1A1*29 seems to have little clinical impact. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28689454-12 2018 Considering the above findings and the current knowledge on UGTs expression in HIM, it is likely that UGT1A10 is mainly responsible for xanthotoxol glucuronidation in the human small intestine, with some contribution from UGT1A1. xanthotoxol 136-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 29891024-10 2018 Previous reports, studies of recombinant UGT isoforms indicated that SN-38 glucuronidation was mainly catalyzed by UGT1A1. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 30673201-4 2018 On the other hand, carriers of various genotypes of UGT1A1 differ significantly in metabolism characteristics of a number of common medications (irinotecan, belinostat, etc. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 30673201-4 2018 On the other hand, carriers of various genotypes of UGT1A1 differ significantly in metabolism characteristics of a number of common medications (irinotecan, belinostat, etc. belinostat 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 29487697-5 2018 Present results support the preemptive screening of mentioned DPYD and UGT1A1 variants to identify patients at risk of clinically relevant 5-fluoruracil- and irinotecan-related AEs, in order to improve treatments" safety through a "genotype-guided" approach. Fluorouracil 139-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29487697-5 2018 Present results support the preemptive screening of mentioned DPYD and UGT1A1 variants to identify patients at risk of clinically relevant 5-fluoruracil- and irinotecan-related AEs, in order to improve treatments" safety through a "genotype-guided" approach. Irinotecan 158-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 29277179-1 2017 BACKGROUND: Uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1), UGT1A1*28 polymorphism can reduce UGT1A1 enzymatic activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 29025858-1 2017 Human UDP-glucuronosyltransferase 1A1 (UGT1A1) is a unique enzyme involved in bilirubin conjugation. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-37 29125289-0 2017 A Practical and High-Affinity Fluorescent Probe for Uridine Diphosphate Glucuronosyltransferase 1A1: A Good Surrogate for Bilirubin. Bilirubin 122-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-99 29125289-2 2017 Several substrates derived from N-butyl-4-phenyl-1,8-naphthalimide were designed and synthesized on the basis of the substrate preference of UGT1A1 and the principle of photoinduced electron transfer (PET). n-butyl-4-phenyl-1,8-naphthalimide 32-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 29125289-4 2017 Both inhibition kinetic analyses and molecular docking simulations demonstrated that 2 could bind on UGT1A1 at the same ligand-binding site as bilirubin. Bilirubin 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29125289-6 2017 In conclusion, a practical and high-affinity fluorescent probe for UGT1A1 was designed and well-characterized, which could serve as a good surrogate for bilirubin to investigate UGT1A1-ligand interactions. Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 29025858-1 2017 Human UDP-glucuronosyltransferase 1A1 (UGT1A1) is a unique enzyme involved in bilirubin conjugation. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 29159025-4 2017 The results demonstrated that both icotinib and erlotinib are UGT1A1 inhibitors, but the inhibitory effect of icotinib on UGT1A1 is weaker than that of erlotinib. icotinib 35-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 29133521-10 2017 A genetic variant at the UGT1A1*28 locus consistently shown to be strongly associated with circulating bilirubin levels-rs6742078-was not significantly associated with blood pressure or hypertension (P>0.05 for all), arguing against a strong causal association of circulating bilirubin with blood pressure. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 28536775-7 2017 Meantime, oroxylin A was rapidly metabolized by UGTs, UGT1A1, -1A3, -1A6, -1A7, -1A8, -1A9, and -1A10 which were involved in the glucuronidation. 5,7-dihydroxy-6-methoxy-2-phenylchromen-4-one 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 28990653-0 2017 Mechanism of in-vitro inhibition of UGT1A1 by paritaprevir. paritaprevir 46-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 28990653-2 2017 Paritaprevir is reported to inhibit human UGT 1A1, but the mechanism of inhibition and its possible clinical consequences are not established. paritaprevir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-49 28990653-3 2017 Our objective was to evaluate the in-vitro metabolic interaction between paritaprevir and the oral contraceptive steroid ethinyl estradiol (EE), a UGT 1A1 substrate. paritaprevir 73-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-154 28990653-3 2017 Our objective was to evaluate the in-vitro metabolic interaction between paritaprevir and the oral contraceptive steroid ethinyl estradiol (EE), a UGT 1A1 substrate. Steroids 113-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-154 28990653-3 2017 Our objective was to evaluate the in-vitro metabolic interaction between paritaprevir and the oral contraceptive steroid ethinyl estradiol (EE), a UGT 1A1 substrate. Ethinyl Estradiol 121-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-154 28990653-9 2017 CONCLUSIONS: Paritaprevir is an in-vitro inhibitor of UGT 1A1. paritaprevir 13-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-61 29159025-0 2017 Comparison of the inhibition potentials of icotinib and erlotinib against human UDP-glucuronosyltransferase 1A1. icotinib 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-111 29159025-0 2017 Comparison of the inhibition potentials of icotinib and erlotinib against human UDP-glucuronosyltransferase 1A1. Erlotinib Hydrochloride 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-111 29159025-4 2017 The results demonstrated that both icotinib and erlotinib are UGT1A1 inhibitors, but the inhibitory effect of icotinib on UGT1A1 is weaker than that of erlotinib. Erlotinib Hydrochloride 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 29159025-3 2017 This study aimed to investigate and compare the inhibitory effects of icotinib and erlotinib against UGT1A1, as well as to evaluate their potential DDI risks via UGT1A1 inhibition. icotinib 70-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29159025-3 2017 This study aimed to investigate and compare the inhibitory effects of icotinib and erlotinib against UGT1A1, as well as to evaluate their potential DDI risks via UGT1A1 inhibition. Erlotinib Hydrochloride 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 29159025-4 2017 The results demonstrated that both icotinib and erlotinib are UGT1A1 inhibitors, but the inhibitory effect of icotinib on UGT1A1 is weaker than that of erlotinib. icotinib 110-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 29159025-5 2017 The IC50 values of icotinib and erlotinib against UGT1A1-mediated NCHN-O-glucuronidation in human liver microsomes (HLMs) were 5.15 and 0.68 mumol/L, respectively. icotinib 19-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 29159025-5 2017 The IC50 values of icotinib and erlotinib against UGT1A1-mediated NCHN-O-glucuronidation in human liver microsomes (HLMs) were 5.15 and 0.68 mumol/L, respectively. Erlotinib Hydrochloride 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 29159025-5 2017 The IC50 values of icotinib and erlotinib against UGT1A1-mediated NCHN-O-glucuronidation in human liver microsomes (HLMs) were 5.15 and 0.68 mumol/L, respectively. nchn-o 66-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 29159025-6 2017 Inhibition kinetic analyses demonstrated that both icotinib and erlotinib were non-competitive inhibitors against UGT1A1-mediated glucuronidation of NCHN in HLMs, with the Ki values of 8.55 and 1.23 mumol/L, respectively. icotinib 51-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 29159025-6 2017 Inhibition kinetic analyses demonstrated that both icotinib and erlotinib were non-competitive inhibitors against UGT1A1-mediated glucuronidation of NCHN in HLMs, with the Ki values of 8.55 and 1.23 mumol/L, respectively. Erlotinib Hydrochloride 64-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 29159025-6 2017 Inhibition kinetic analyses demonstrated that both icotinib and erlotinib were non-competitive inhibitors against UGT1A1-mediated glucuronidation of NCHN in HLMs, with the Ki values of 8.55 and 1.23 mumol/L, respectively. nchn 149-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 29159025-8 2017 These findings are helpful for the medicinal chemists to design and develop next generation tyrosine kinase inhibitors with improved safety, as well as to guide reasonable applications of icotinib and erlotinib in clinic, especially for avoiding their potential DDI risks via UGT1A1 inhibition. icotinib 188-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 276-282 29159025-8 2017 These findings are helpful for the medicinal chemists to design and develop next generation tyrosine kinase inhibitors with improved safety, as well as to guide reasonable applications of icotinib and erlotinib in clinic, especially for avoiding their potential DDI risks via UGT1A1 inhibition. Erlotinib Hydrochloride 201-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 276-282 28934153-9 2017 Selaginellin and selaginellin M also showed medium inhibitory potential against CYP2C9, CYP2J2, UGT1A1, and UGT1A3 (1 muM < IC50 < 5 muM). selaginellin S 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 29113203-1 2017 Uridine diphosphate glucuronosyltransferase 1A (UGT1A1), which affects irinotecan metabolism, has been associated with severe adverse reactions in patients with cancer treated with irinotecan. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-1 2017 Uridine diphosphate glucuronosyltransferase 1A (UGT1A1), which affects irinotecan metabolism, has been associated with severe adverse reactions in patients with cancer treated with irinotecan. Irinotecan 181-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 29113203-9 2017 These findings suggested that the UGT1A1*28 and UGT1A1*6 genotypes may be associated with irinotecan-induced severe toxicity, and clarified the clinical importance of UGT1A1 polymorphisms, particularly UGT1A1*6, regarding irinotecan therapy in Chinese patients. Irinotecan 222-232 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 29072417-1 2017 Irinotecan life threatening toxicity is partly related to cytotoxic drugmetabolite which is primarily inactivated by the UGT1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 29072417-1 2017 Irinotecan life threatening toxicity is partly related to cytotoxic drugmetabolite which is primarily inactivated by the UGT1A1 enzyme. drugmetabolite 68-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 29072417-2 2017 The primary aim of the present research was tofind the correlation between UGT1A1-genotype and clinical toxicity of irinotecan. Irinotecan 116-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 29072417-13 2017 Conclusions: UGT1A1 28*/28* is strongly associatedwith drug"s life-threatening toxicity even in a moderate dose of Irinotecan. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 28934153-9 2017 Selaginellin and selaginellin M also showed medium inhibitory potential against CYP2C9, CYP2J2, UGT1A1, and UGT1A3 (1 muM < IC50 < 5 muM). selaginellin M 17-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 28932176-2 2017 The present research was aimed to study the effect of 2 important single nucleotide polymorphisms (SNPs; rs8330 and rs10929303) of UGT1A1 gene on glucuronidation status of acetaminophen in healthy volunteers (n = 109). Acetaminophen 172-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 28493308-9 2017 CONCLUSIONS: UGT1A1 genotype-directed dosing (gCAPIRINOX) appears to be feasible with favorable rates of hematologic toxicity compared with prior 3-drug studies in unselected patients. gcapirinox 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 28915895-0 2017 Impact of UGT1A1 gene polymorphisms on plasma dolutegravir trough concentrations and neuropsychiatric adverse events in Japanese individuals infected with HIV-1. dolutegravir 46-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 28915895-1 2017 BACKGROUND: Dolutegravir (DTG) is metabolized mainly by uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1), and partly by cytochrome P450 3A (CYP3A). dolutegravir 12-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-109 28915895-1 2017 BACKGROUND: Dolutegravir (DTG) is metabolized mainly by uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1), and partly by cytochrome P450 3A (CYP3A). dolutegravir 12-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 28915895-1 2017 BACKGROUND: Dolutegravir (DTG) is metabolized mainly by uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1), and partly by cytochrome P450 3A (CYP3A). dolutegravir 26-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-109 28915895-1 2017 BACKGROUND: Dolutegravir (DTG) is metabolized mainly by uridine diphosphate (UDP)-glucuronosyltransferase 1A1 (UGT1A1), and partly by cytochrome P450 3A (CYP3A). dolutegravir 26-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 28915895-10 2017 Multivariate analysis revealed that carrying one or two UGT1A1*6 gene polymorphisms, one UGT1A1*28 polymorphism, and age of < 40 years were independent factors associated with high DTG trough concentrations. dolutegravir 184-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 28915895-10 2017 Multivariate analysis revealed that carrying one or two UGT1A1*6 gene polymorphisms, one UGT1A1*28 polymorphism, and age of < 40 years were independent factors associated with high DTG trough concentrations. dolutegravir 184-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 28915895-13 2017 CONCLUSION: In addition to younger age, carrying UGT1A1*6 and/or UGT1A1*28 was demonstrated to be a factor associated with high DTG trough concentrations. dolutegravir 128-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 28915895-13 2017 CONCLUSION: In addition to younger age, carrying UGT1A1*6 and/or UGT1A1*28 was demonstrated to be a factor associated with high DTG trough concentrations. dolutegravir 128-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 28932176-4 2017 The in vivo activity of UGT1A1 was investigated by high-performance liquid chromatography-based analysis of glucuronidation status (ie, acetaminophen and acetaminophen glucuronide) in human volunteers after oral intake of a single dose (1000 mg) of acetaminophen. Acetaminophen 136-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28932176-4 2017 The in vivo activity of UGT1A1 was investigated by high-performance liquid chromatography-based analysis of glucuronidation status (ie, acetaminophen and acetaminophen glucuronide) in human volunteers after oral intake of a single dose (1000 mg) of acetaminophen. acetaminophen glucuronide 154-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28932176-4 2017 The in vivo activity of UGT1A1 was investigated by high-performance liquid chromatography-based analysis of glucuronidation status (ie, acetaminophen and acetaminophen glucuronide) in human volunteers after oral intake of a single dose (1000 mg) of acetaminophen. Acetaminophen 154-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28474802-0 2017 Rapid and sensitive detection of UGT1A1 polymorphisms associated with irinotecan toxicity by a novel DNA microarray. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28552915-13 2017 This study demonstrates that UGT2B7 common SNPs and their dimers with UGT1A1 and UGT1A9 and their allelic variants can regioselectively affect the generation of two metabolites of morphine via altering the CLint ratios of M3G to M6G. Morphine 180-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 28567595-0 2017 Do Alpha Thalassemia, Fetal Hemoglobin, and the UGT1A1 Polymorphism have an Influence on Serum Bilirubin Levels and Cholelithiasis in Patients with Sickle Cell Disease? Bilirubin 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 28567595-2 2017 OBJECTIVES: The objective of this study was to determine the combined influence of alpha thalassemia, fetal hemoglobin, and the UGT1A1 polymorphism on serum bilirubin levels and cholelithiasis in patients with sickle cell disease. Bilirubin 157-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 28567595-6 2017 Bilirubin levels were influenced by the UGT1A1 polymorphism but not by alpha thalassemia and fetal hemoglobin. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 28567595-8 2017 CONCLUSION: These preliminary findings suggest that the UGT1A1 gene can influence serum bilirubin levels in sickle cell anemia and serve as a tool to differentiate an acute hemolytic condition from a pre-existing condition of hyperbilirubinemia. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 28603997-0 2017 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by Polyphenolic Acids Impact Safety of Popular Salvianolic Acid A/B-Containing Drugs and Herbal Products. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28603997-0 2017 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by Polyphenolic Acids Impact Safety of Popular Salvianolic Acid A/B-Containing Drugs and Herbal Products. polyphenolic acids 65-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28603997-0 2017 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by Polyphenolic Acids Impact Safety of Popular Salvianolic Acid A/B-Containing Drugs and Herbal Products. salvianolic acid A 109-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28603997-0 2017 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by Polyphenolic Acids Impact Safety of Popular Salvianolic Acid A/B-Containing Drugs and Herbal Products. Boron 36-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28603997-3 2017 The purpose of this article is to determine the mechanism by which certain polyphenolic acids inhibit UGT1A1-mediated bilirubin glucuronidation, leading to jaundice or hyperbilirubinemia. polyphenolic acids 75-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 28603997-3 2017 The purpose of this article is to determine the mechanism by which certain polyphenolic acids inhibit UGT1A1-mediated bilirubin glucuronidation, leading to jaundice or hyperbilirubinemia. Bilirubin 118-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 28603997-6 2017 SAA, SAB, DSI, and CDI, but not DSS, PA, and RA, significantly inhibited human UGT1A1-mediated bilirubin glucuronidation via a mixed-type inhibitory mechanism. salvianolic acid B 5-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 28603997-6 2017 SAA, SAB, DSI, and CDI, but not DSS, PA, and RA, significantly inhibited human UGT1A1-mediated bilirubin glucuronidation via a mixed-type inhibitory mechanism. N-hydroxysuccinimide suberic acid ester 10-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 28603997-6 2017 SAA, SAB, DSI, and CDI, but not DSS, PA, and RA, significantly inhibited human UGT1A1-mediated bilirubin glucuronidation via a mixed-type inhibitory mechanism. Bilirubin 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 28654297-7 2017 Recombinant human uridine-5"-diphospho -glucuronosyltransferase (UGT) isoforms and chemical inhibition studies demonstrated that UGT1As (mainly UGT1A1, UGT1A9, UGT1A10) and UGT2B7 were likely the main contributors to eriodictyol glucuronidation. eriodictyol 217-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-133 28397089-9 2017 CONCLUSION: The VCS confirmed the importance of genetic polymorphisms of UGT1A1 *28 and SLCO1B1 c.521T>C in the irinotecan induced side effects. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28585035-0 2017 UGT1A1*6 polymorphisms are correlated with irinotecan-induced neutropenia: a systematic review and meta-analysis. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28474802-4 2017 It is established that the polymorphisms of UGT1A1*28 and UGT1A1*6 are significantly associated with severe toxicity induced by the anti-cancer drug irinotecan. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 28474802-4 2017 It is established that the polymorphisms of UGT1A1*28 and UGT1A1*6 are significantly associated with severe toxicity induced by the anti-cancer drug irinotecan. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 28157715-0 2017 Combined use of irinotecan with histone deacetylase inhibitor belinostat could cause severe toxicity by inhibiting SN-38 glucuronidation via UGT1A1. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 28074472-0 2017 Cost Evaluation of Irinotecan-Related Toxicities Associated With the UGT1A1*28 Patient Genotype. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 28074472-1 2017 The adoption of a preemptive UGT1A1*28 genotyping to increase irinotecan safety in clinical practice is still limited. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 28892962-2 2017 It is caused by mutations in the UGT1A1 gene which codes for the enzyme uridine diphosphate glucoronosyl transferase- 1, required for the conjugation and further excretion of bilirubin from the body. Bilirubin 175-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28637434-11 2017 In the first-line irinotecan-based treatment, UGT1A1*28 and DPYD*5 contributed to higher response rates (P = 0.043 and P = 0.019, respectively), while DPYD*5 was found to associate with better progression-free survival (P = 0.015). Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 28637434-13 2017 CONCLUSION: Results still showed UGT1A1*6 and UGT1A1*28 to be partially associated with irinotecan-induced toxicity and clinical response. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 28637434-13 2017 CONCLUSION: Results still showed UGT1A1*6 and UGT1A1*28 to be partially associated with irinotecan-induced toxicity and clinical response. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 28804517-8 2017 It is imperative to check UGT1A1 gene status in all patients being considered for treatment with nanoliposomal irinotecan. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28157715-0 2017 Combined use of irinotecan with histone deacetylase inhibitor belinostat could cause severe toxicity by inhibiting SN-38 glucuronidation via UGT1A1. belinostat 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 28157715-4 2017 The data showed that belinostat at 100 and 200 micromol/L inhibited SN-38 glucuronidation via UGT1A1 in a dose-dependent manner, causing significant decrease in Vmax and CLint (p < 0.05) from 12.60 to 1.95 pmol/min/mg and 21.59 to 4.20 muL/min/mg protein respectively. belinostat 21-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 28157715-4 2017 The data showed that belinostat at 100 and 200 micromol/L inhibited SN-38 glucuronidation via UGT1A1 in a dose-dependent manner, causing significant decrease in Vmax and CLint (p < 0.05) from 12.60 to 1.95 pmol/min/mg and 21.59 to 4.20 muL/min/mg protein respectively. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 28790841-0 2017 Relationship between UGT1A1*6/*28 gene polymorphisms and the efficacy and toxicity of irinotecan-based chemotherapy. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 28790862-6 2017 Pharmacokinetics for additional HIV drugs, such as the integrase inhibitors Raltegravir and Elvitegravir, has been also shown to be influenced by UGT1A1 polymorphisms. Raltegravir Potassium 76-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 28790862-6 2017 Pharmacokinetics for additional HIV drugs, such as the integrase inhibitors Raltegravir and Elvitegravir, has been also shown to be influenced by UGT1A1 polymorphisms. elvitegravir 92-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-128 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 28790841-1 2017 PURPOSE: A retrospective study was performed to analyze the relationship between uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *6/*28 gene polymorphisms and adverse reactions associated with irinotecan (CPT-11)-based chemotherapy. Irinotecan 214-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-128 28790841-2 2017 The correlation between UGT1A1 polymorphisms and the clinical efficacy of CPT-11 was also analyzed, along with the influence of age and tumor type. Irinotecan 74-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 28790841-5 2017 RESULTS: Colorectal cancer patients with the UGT1A1*6 mutant genotype had a significantly higher risk of severe delayed diarrhea than that of wild-type individuals when administered a CPT-11 dose >=130 mg/m2 (P=0.042); the same phenomenon was observed when the UGT1A1*6 and UGT1A1*28 mutant genotypes were considered together (P=0.028). Irinotecan 184-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 28790841-6 2017 However, in lung cancer patients administered a low dose of CPT-11, UGT1A1*6/*28 variants were not significantly associated with severe neutropenia or delayed diarrhea. Irinotecan 60-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28213806-4 2017 The presence of two UGT1A1 variants (consistent with Gilbert or Crigler-Najjar syndrome) occurred less frequently in neonates (aged <=28 days) than older children (aged 1-18 years) (31.3% in neonates vs. 85.1%, p < 0.0001), and among neonates there was no significant difference in mean total bilirubin between those with two UGT1A1 variants and those without (p = 0.47). Bilirubin 299-308 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28283499-8 2017 In addition to NGMN, we have also elucidated the biotransformation of norgestrel (NG), a downstream norgestimate and NGMN metabolite, and found that CYP3A4 and UGT1A1 have a major contribution to the elimination of NG with a combined fm value of 1. Norgestrel 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 28283499-8 2017 In addition to NGMN, we have also elucidated the biotransformation of norgestrel (NG), a downstream norgestimate and NGMN metabolite, and found that CYP3A4 and UGT1A1 have a major contribution to the elimination of NG with a combined fm value of 1. norgestimate 100-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 28502040-0 2017 UGT1A1 polymorphisms with irinotecan-induced toxicities and treatment outcome in Asians with Lung Cancer: a meta-analysis. Irinotecan 26-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28502040-1 2017 Previous studies of irinotecan pharmacogenetics have shown that the UGT1A1*28 polymorphism has an effect on irinotecan (IRI)-induced toxicities in Caucasians. Irinotecan 120-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 28246004-4 2017 Among them, UGT1A1, UGT1A3, UGT1A6, UGT1A9 and UGT2B7 contributed 95.6% to genistein glucuronidation in human liver, with significant correlation between gene expression of the five UGTs and the glucuronidation of genistein. Genistein 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28242239-9 2017 Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. Paclitaxel 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 28456840-5 2017 CNSDose (ABCB1, ABCC1, CYP2C19, CYP2D6, UGT1A1) is the only pharmacogenomics test that significantly improved remission (one additional remitting patient in 12 weeks per three genotyped, 95% CI 1.7 to 3.5) and reduced intolerability in an RCT. cnsdose 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 28242239-9 2017 Paclitaxel-mediated induction of genes involved in irinotecan metabolism such as Cyp3a11 and Ugt1a1 was TLR4-dependent, while induction of the transporter Mrp2 was TLR4-independent. Irinotecan 51-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 28283555-3 2017 Although neonatal jaundice is mostly benign, excessively high levels of serum bilirubin in a small percentage of newborns can cause bilirubin-induced neurologic dysfunction, potentially leading to permanent brain damage, a condition known as kernicterus Although a large portion of hyperbilirubinemia cases in newborns are associated with hemolytic diseases, we emphasize here the impaired ability of UGT1A1 to eliminate bilirubin that contributes to hyperbilirubinemia-induced neurotoxicity in the developmental stage. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 401-407 27938508-0 2017 Regorafenib Plus FOLFIRI With Irinotecan Dose Escalated According to Uridine Diphosphate Glucuronosyltransferase 1A1 Genotyping in Patients With Metastatic Colorectal Cancer. regorafenib 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-116 27938508-0 2017 Regorafenib Plus FOLFIRI With Irinotecan Dose Escalated According to Uridine Diphosphate Glucuronosyltransferase 1A1 Genotyping in Patients With Metastatic Colorectal Cancer. FOLFIRI regimen 17-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-116 27938508-0 2017 Regorafenib Plus FOLFIRI With Irinotecan Dose Escalated According to Uridine Diphosphate Glucuronosyltransferase 1A1 Genotyping in Patients With Metastatic Colorectal Cancer. Irinotecan 30-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-116 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. regorafenib 27-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. regorafenib 27-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. regorafenib 27-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. FOLFIRI regimen 44-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. FOLFIRI regimen 44-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. FOLFIRI regimen 44-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 27938508-8 2017 Our findings indicate that regorafenib plus FOLFIRI with irinotecan dose escalation based on UGT1A1 genotyping in previously treated patients with mCRC and with UGT1A1*1/*1 and UGT1A1*1/*28 genotypes is clinically effective and yields improved oncological outcomes. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 28283555-2 2017 Owing to the fact that bilirubin is metabolized solely through glucuronidation by UDP-glucuronosyltransferase (UGT) 1A1, it is now known that immaturity of UGT1A1, in combination with the overproduction of bilirubin during the developmental stage, acts as a bottleneck to bilirubin elimination and predisposes the infant to high total serum bilirubin levels. Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-119 28283555-2 2017 Owing to the fact that bilirubin is metabolized solely through glucuronidation by UDP-glucuronosyltransferase (UGT) 1A1, it is now known that immaturity of UGT1A1, in combination with the overproduction of bilirubin during the developmental stage, acts as a bottleneck to bilirubin elimination and predisposes the infant to high total serum bilirubin levels. Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 28283555-3 2017 Although neonatal jaundice is mostly benign, excessively high levels of serum bilirubin in a small percentage of newborns can cause bilirubin-induced neurologic dysfunction, potentially leading to permanent brain damage, a condition known as kernicterus Although a large portion of hyperbilirubinemia cases in newborns are associated with hemolytic diseases, we emphasize here the impaired ability of UGT1A1 to eliminate bilirubin that contributes to hyperbilirubinemia-induced neurotoxicity in the developmental stage. Bilirubin 132-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 401-407 28283555-3 2017 Although neonatal jaundice is mostly benign, excessively high levels of serum bilirubin in a small percentage of newborns can cause bilirubin-induced neurologic dysfunction, potentially leading to permanent brain damage, a condition known as kernicterus Although a large portion of hyperbilirubinemia cases in newborns are associated with hemolytic diseases, we emphasize here the impaired ability of UGT1A1 to eliminate bilirubin that contributes to hyperbilirubinemia-induced neurotoxicity in the developmental stage. Bilirubin 132-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 401-407 28096081-1 2017 Gilbert"s syndrome in humans is derived from a polymorphism (TA repeat) in the hepatic UGT1A1 gene that results in decreased conjugation and increased levels of unconjugated bilirubin. Bilirubin 174-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 27359323-7 2017 Vitamin A exerted competitive inhibition on the activity of UGT1A1, 2B4, 2B7, and 2B15, and the inhibition kinetic parameters (Ki) were calculated to be 31.1, 16.8, 2.2, and 11.6 muM for UGT1A1, 2B4, 2B7, and 2B15. Vitamin A 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 27359323-7 2017 Vitamin A exerted competitive inhibition on the activity of UGT1A1, 2B4, 2B7, and 2B15, and the inhibition kinetic parameters (Ki) were calculated to be 31.1, 16.8, 2.2, and 11.6 muM for UGT1A1, 2B4, 2B7, and 2B15. Vitamin A 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 27977017-1 2017 OBJECTIVE: The objective of our study was to measure the effect of genetic variants of these two enzymes, UGT1A1 and SLCO1B1, in the bilirubin metabolic pathway on the degree of hyperbilirubinemia in a cohort of African-American (AA) infants from our well-baby nursery. Bilirubin 133-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Fluorouracil 112-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Mercaptopurine 128-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 27641154-7 2017 Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 27832386-3 2017 While polymorphism of UDP-glucuronosyltransferase1A1 (UGT1A1) is involved in detoxification of benzo(a)pyrene-7,8-dihydrodiol(-), a major tobacco carcinogen, the association between UGT1A1 genotype and lung cancer has not been examined. benzo(a)pyrene 7,8-dihydrodiol 95-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 27832386-3 2017 While polymorphism of UDP-glucuronosyltransferase1A1 (UGT1A1) is involved in detoxification of benzo(a)pyrene-7,8-dihydrodiol(-), a major tobacco carcinogen, the association between UGT1A1 genotype and lung cancer has not been examined. benzo(a)pyrene 7,8-dihydrodiol 95-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 28065859-7 2017 LAP, PAZ, REG and SOR inhibited UGT1A1-catalysed bilirubin glucuronidation with mean IC50 values ranging from 34nM (REG) to 3734nM (PAZ). Bilirubin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 28296739-0 2017 UGT1A1*28 relationship with abnormal total bilirubin levels in chronic hepatitis C patients: Outcomes from a case-control study. Bilirubin 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28346059-5 2017 UGT1A1 rs887829TT (p = 0.002) and CYP1A2 rs762551CC (p = 0.019) resulted as predictive factor of ferritin levels and CYP1A1 rs2606345CA/AA (p = 0.021) and CYP1A2 rs762551AC/CC (p = 0.027) of liver iron concentration. Iron 197-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27180825-15 2017 Glucuronidation of belinostat was markedly inhibited by emodin and apigenin (two potent inhibitors of UGT1A1), and by quinidine and diclofenac sodium (two selective inhibitors of UGT2B7). belinostat 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 28446312-5 2017 Further gene evaluation disclosed a insertion mutation in the (TA)6TAA box, and a missense mutation(G A) at 211 bp of exon 1, corresponding to the deficiency in the bilirubin-conjugating enzyme uridine-diphosphoglucuronosyl transferase1A1 (UGT1A1). Bilirubin 165-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-238 28446312-5 2017 Further gene evaluation disclosed a insertion mutation in the (TA)6TAA box, and a missense mutation(G A) at 211 bp of exon 1, corresponding to the deficiency in the bilirubin-conjugating enzyme uridine-diphosphoglucuronosyl transferase1A1 (UGT1A1). Bilirubin 165-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 240-246 28263463-7 2017 The data show that this drug inhibits the enzyme UDP-glucuronosyl transferase-1A1, responsible for conjugating bilirubin with glucuronic acid. Bilirubin 111-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-81 28263463-7 2017 The data show that this drug inhibits the enzyme UDP-glucuronosyl transferase-1A1, responsible for conjugating bilirubin with glucuronic acid. Glucuronic Acid 126-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-81 28338110-3 2017 The UGT1A1 promoter (TA) repeats variants are documented of being involved in abnormally elevated bilirubin levels. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 28338110-4 2017 The aim of the present study is to analyze the impact of UGT1A1 promoter variants on bilirubin levels in Romanian patients clinically supected with GS. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 28296739-11 2017 Total bilirubin increase was noticed according to thymine-adenine repeats in genotypes (P < 0.001), and the TB greater than 1 mg/dL group had more UGT1A1*28 subjects than in the group with TB values <1 mg/dL (18.3 vs 5.3; P < 0.001). Bilirubin 111-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 28296739-13 2017 Among patients with increased TB levels, the frequency of UGT1A1*28 is higher than those with normal TB. Bilirubin 30-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 28296739-13 2017 Among patients with increased TB levels, the frequency of UGT1A1*28 is higher than those with normal TB. Bilirubin 101-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 28278081-0 2017 Correlative analysis of plasma SN-38 levels and DPD activity with outcomes of FOLFIRI regimen for metastatic colorectal cancer with UGT1A1 *28 and *6 wild type and its implication for individualized chemotherapy. Irinotecan 31-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 28278081-1 2017 It remains uncertain whether there is an correlation between clinical pharmacokinetic parameters and outcomes for metastatic colorectal cancer especially with UGT1A1 *28 and *6 wild type (*1/*1-*1/*1) for serious events associated with Irinotecan are largely excluded. Irinotecan 236-246 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 28278081-2 2017 This study retrospectively analyzed the relationship between Irinotecan metabolite levels and outcomes of UGT1A1 *1/*1-*1/*1 genotype arrangement. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 28278081-8 2017 Therefore, plasma SN-38 levels is related to outcomes for UGT1A1 *1/*1-*1/*1 genotype, to improve efficacy, patients with CSN-38 1.5 h lower than 50.24 ng/ml, CPT-11 dosage could be added in next chemmotherapy on SN-38 plasma level monitoring. Irinotecan 18-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 27915055-7 2017 The results indicated that UGT1A1 and UGT1A9 were the main isoforms involved in the formation of isofraxidin-7-O-glucuronide. isofraxidin-7-o-glucuronide 97-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 28296739-4 2017 Our aim was to assess UGT1A1 genotypes" frequency in chronic hepatitis C (CHC) patients and correlate with total bilirubin (TB). Bilirubin 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 28296739-4 2017 Our aim was to assess UGT1A1 genotypes" frequency in chronic hepatitis C (CHC) patients and correlate with total bilirubin (TB). Bilirubin 124-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 28280378-0 2017 UGT1A1 polymorphisms in cancer: impact on irinotecan treatment. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 175-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 175-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 182-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 182-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 233-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 28280378-3 2017 Among them, UGT1A1*28 and UGT1A1*6 have been reported to be associated with severe toxicities in patients treated with irinotecan-based chemotherapy by increasing the dose of SN-38 (7-ethyl-10-hydroxycamptothecin), an active form of irinotecan. Irinotecan 233-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 28280378-4 2017 Many association studies and meta-analyses have demonstrated the contribution of UGT1A1*28 and UGT1A1*6 polymorphisms to the toxicities caused by irinotecan-based therapy. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 28280378-4 2017 Many association studies and meta-analyses have demonstrated the contribution of UGT1A1*28 and UGT1A1*6 polymorphisms to the toxicities caused by irinotecan-based therapy. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 28131654-2 2017 We investigated whether the polymorphisms of UGT1A1 and UGT1A3 played an important role in the disposition of the drug after oral administration of febuxostat tablet in Chinese subjects. Febuxostat 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 27507617-1 2017 Purpose:UGT1A1*28 confers a higher risk of toxicity in patients treated with irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 27507617-8 2017 Changes in the AUCs of irinotecan and SN-38 associated with bevacizumab treatment were marginal.Conclusions: The MTD of irinotecan in FOLFIRI plus bevacizumab is 310 mg/m2 for UGT1A1 *1/*1 patients and 260 mg/m2 for *1/*28 patients. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 28173214-0 2017 UGT1A1 polymorphism as a prognostic indicator of stage I ovarian clear cell carcinoma patients treated with irinotecan. Irinotecan 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 28173214-1 2017 We investigated whether UGT1A1 polymorphisms are associated with the prognosis of ovarian cancer patients treated with irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 28173214-7 2017 We conclude that UGT1A1 polymorphisms have the potential to be a prognostic marker of irinotecan treatment. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 28017536-5 2017 Glucuronidation of a carboxylate residue is catalyzed by UDP-glucuronosyltransferase 1A and 2B isoforms. carboxylate 21-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-94 28367249-0 2017 Predictive Value of UGT1A1*28 Polymorphism In Irinotecan-based Chemotherapy. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 28367249-1 2017 The UGT1A1*28 polymorphism was suggested to be significantly connected with irinotecan-induced toxicity and response to chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 28367249-3 2017 Hence we carried out a meta-analysis to investigate the effect of UGT1A1*28 polymorphism on severe diarrhea, neutropenia, and response of patients who had undergone irinotecan-based chemotherapy. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 28367249-10 2017 Our data showed that the UGT1A1*28 polymorphism had a significant relationship with toxicity and response to irinotecan-based chemotherapy. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 27654129-3 2017 Dose escalation of irinotecan was performed according to the results of UGT1A1 genotyping in all patients. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 29956901-11 2017 CONCLUSION: SXD could significantly reduce irinotecan-induced hematological and gastrointestinaltoxicities in UGT1A1*28 or *6 variant patients (high risk group), while this treatment didn"t affect clinicalresponse of chemotherapy. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 29098099-0 2017 Life-Threatening Irinotecan-Induced Toxicity in an Adult Patient with Alveolar Rhabdomyosarcoma: The Role of a UGT1A1 Polymorphism. Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-67 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 29098099-7 2017 The analysis of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene revealed homozygous UGT1A1 *28 polymorphism with an associated homozygous mutation c.-3275T>G; the latter is associated with a decrease of about 80% of UGT1A1 transcription explaining this irinotecan induced toxicity. Irinotecan 274-284 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 29098099-8 2017 Physician must be aware of the potential hematological (mainly neutropenia and infectious disease) and digestive (mainly diarrhea) toxicities caused by irinotecan and especially when the patient presents a UGT1A1 *28 homozygous allele. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 206-212 28395759-2 2017 Patients were stratified by their pharmacogenetic/phenotypic status: the irinotecan dose was adjusted according to the number of TA tandem repeats in the UGT1A1 promoter, while the 5-fluorouracil (5-FU) dose was initially adjusted according to dihydropyrimidine dehydrogenase (DPD) activity at initial screening (5-FUODPM Tox) followed by PK-guided dose optimization (5-FUODPM Protocol). Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 28952244-2 2017 Therefore, changes in UGT1A1 expression/functional can not only cause adverse clinical drug/herbs-drug interactions, but also lead to metabolic disorder of endogenous substances, causing high blood bilirubin, bilirubin encephalopathy and liver injury, as well as other side effects. Bilirubin 198-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 28952244-2 2017 Therefore, changes in UGT1A1 expression/functional can not only cause adverse clinical drug/herbs-drug interactions, but also lead to metabolic disorder of endogenous substances, causing high blood bilirubin, bilirubin encephalopathy and liver injury, as well as other side effects. Bilirubin 209-218 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 28952244-4 2017 This article would summarize the advances in research on drug metabolism and toxicology in domestic and foreign literature, and investigate the regulatory effects of different types of traditional Chinese medicine(TCM) ingredients(such as flavonoids, coumarins, alkaloids) on UGT1A1 expression and activity, including inhibitory effect of TCM chemical ingredients on UGT1A1 and effect of TCM chemical ingredients on UGT1A1. Alkaloids 262-271 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 276-282 28098838-0 2017 Role of UDP-Glucuronosyltransferase 1A1 in the Metabolism and Pharmacokinetics of Silymarin Flavonolignans in Patients with HCV and NAFLD. Silymarin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 28098838-0 2017 Role of UDP-Glucuronosyltransferase 1A1 in the Metabolism and Pharmacokinetics of Silymarin Flavonolignans in Patients with HCV and NAFLD. Flavonolignans 92-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 164-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 175-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 183-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 194-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. flavonolignan glucuronide 270-295 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 413-415 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-3 2017 The aims of this investigation were: (1) to determine the association of UGT1A1*28 polymorphism with the area under the plasma concentration-time curves (AUCs) for silybin A (SA) and silybin B (SB); (2) to evaluate the effect of UGT1A1*28 polymorphism on the profile of flavonolignan glucuronide conjugates found in the plasma; and (3) to investigate the role of UGT1A1 enzyme kinetics on the pharmacokinetics of SA and SB. Silybin 420-422 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 28098838-6 2017 In contrast, an increase in the proportion of sulfated flavonolignan conjugates in plasma was observed in subjects with UGT1A1*28/*28 genotype compared to subjects carrying wild type alleles. Flavonolignans 55-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 28098838-7 2017 Differences in SA and SB in vitro intrinsic clearance estimates for UGTIA1 correlated inversely with SA and SB exposures observed in vivo indicating a major role for UGT1A1 in silymarin metabolism. Silymarin 176-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 28098838-8 2017 In addition, a significant difference in the metabolic ratio observed between patients with NAFLD and HCV suggests that any effect of UGT1A1 polymorphism may be obscured by a greater effect of liver disease on the pharmacokinetics of silymarin. Silymarin 234-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 28399191-10 2017 The UGT1A1*28 polymorphism was detected more often among neonates with elevated bilirubin. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 27943244-1 2017 OBJECTIVE: The present study was undertaken to investigate the genotype and allele frequencies of the variants in the four bilirubin metabolism genes (UGT1A1, OATP2, HMOX1, and BLVRA) and their association with hyperbilirubinemia. Bilirubin 123-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 27654129-7 2017 FOLFIRI plus bevacizumab as a first-line chemotherapy may achieve effective disease control and be safe in patients with mCRC and hyperbilirubinemia based on UGT1A1 genotyping. FOLFIRI regimen 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 27503581-5 2017 Both UGT1A1*6 and *28 were reliably demonstrated to be risk factors for irinotecan-induced neutropenia, with tests for both polymorphisms potentially being particularly useful in Asian cancer patients. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 27840292-9 2017 Metabolism and transport of both HT and FA were curtailed by the organic acid lactate owing to a reduction of UGT1A1 protein levels. organic acid lactate 65-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 27908259-1 2017 BACKGROUND: Bilirubin is a toxic waste product of metabolism, eliminated mainly through UGT1A1 mediated conjugation to mono- and di-glucuronides. Bilirubin 12-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 27908259-1 2017 BACKGROUND: Bilirubin is a toxic waste product of metabolism, eliminated mainly through UGT1A1 mediated conjugation to mono- and di-glucuronides. mono- and di-glucuronides 119-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 27908259-8 2017 The Km and Vmax values for total bilirubin glucuronide formations were determined to be 0.05 +- 0.01 muM and 181.9 +- 5.3 pmol/min/mg-protein, respectively, in human recombinant UGT1A1, and 0.23 +- 0.05 muM and 875 +- 45 pmol/min/mg protein in human liver microsomes (HLM). Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-184 27908259-8 2017 The Km and Vmax values for total bilirubin glucuronide formations were determined to be 0.05 +- 0.01 muM and 181.9 +- 5.3 pmol/min/mg-protein, respectively, in human recombinant UGT1A1, and 0.23 +- 0.05 muM and 875 +- 45 pmol/min/mg protein in human liver microsomes (HLM). Glucuronides 43-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-184 27704169-1 2017 PURPOSE: Complete or partial inactivity of UGT1A1, the unique enzyme responsible for bilirubin glucuronidation, is commonly associated with hyperbilirubinemia. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 28951772-0 2017 Serum Bilirubin Levels and Promoter Variations in HMOX1 and UGT1A1 Genes in Patients with Fabry Disease. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 28951772-7 2017 However, the presence of the TA7 allele UGT1A1 gene promoter, responsible for higher systemic bilirubin levels, was associated with a twofold lower risk of manifestation of FD (OR = 0.51, 95% CI = 0.27-0.97, p = 0.038). Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 27967321-0 2017 The impact of the UGT1A1*60 allele on bilirubin serum concentrations. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 27967321-2 2017 MATERIALS & METHODS: Retrospective review of clinically obtained serum bilirubin concentrations in pediatric patients to evaluate the association of the UGT1A1 -3279T>G (*60) variant with bilirubin concentrations and assessed linkage disequilibrium of the UGT1A1 -3279T>G (*60) and A(TA)7TAA (*28) variants. Bilirubin 195-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 26999266-13 2017 Breast mRNA expression of UGTs capable of glucuronidating bisphenol-A was highest for UGT1A1, followed by UGT2B4, UGT1A9, UGT1A10, UGT2B7 and UGT2B15. bisphenol A 58-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 27967321-4 2017 Total bilirubin concentration was lower in patients homozygous for the UGT1A1 -3279T>G (*60/*60) variant than in patients homozygous for the UGT1A1 A(TA)7TAA (*28/*28) variant (p < 0.01). Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 27967321-4 2017 Total bilirubin concentration was lower in patients homozygous for the UGT1A1 -3279T>G (*60/*60) variant than in patients homozygous for the UGT1A1 A(TA)7TAA (*28/*28) variant (p < 0.01). Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 27883280-0 2017 Phase I study of irinotecan for previously treated lung cancer patients with the UGT1A1*28 or *6 polymorphism: Results of the Lung Oncology Group in Kyushu (LOGIK1004A). Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-90 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-98 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-90 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-98 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27883280-1 2017 BACKGROUND: Various polymorphisms have been detected in the UDP-glucuronosyltransferase 1A ( UGT1A ) gene, and UGT1A1 *28 and UGT1A1 *6 have important effects on the pharmacokinetics of irinotecan and the risk of severe toxicities during irinotecan therapy. Irinotecan 238-248 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 27883280-2 2017 This study was conducted to determine the maximum tolerated dose (MTD) of irinotecan chemotherapy according to the UGT1A1 genotype in previously treated lung cancer patients with the UGT1A1 *28 or UGT1A1 *6 polymorphism. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 27883280-10 2017 CONCLUSIONS: The MTD of irinotecan for previously treated lung cancer patients that are heterozygous for the UGT1A1 * 28 or UGT1A1 * 6 gene polymorphism is 60 mg/m2 . Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 27883280-10 2017 CONCLUSIONS: The MTD of irinotecan for previously treated lung cancer patients that are heterozygous for the UGT1A1 * 28 or UGT1A1 * 6 gene polymorphism is 60 mg/m2 . Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 26999266-14 2017 Bisphenol-A glucuronidation was over 10-fold lower in breast tissues with the UGT1A1*28 allele compared with tissues without this allele (p = 0.006). bis(4-hydroxyphenyl)sulfone 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 27704562-3 2016 Since raltegravir is eliminated mainly by metabolism via an UDP-glucuronosyltransferase (UGT) 1 A1-mediated glucuronidation pathway, co-administration of UGT1A1 inducers may alter plasma levels of raltegravir. Raltegravir Potassium 6-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-98 27704562-3 2016 Since raltegravir is eliminated mainly by metabolism via an UDP-glucuronosyltransferase (UGT) 1 A1-mediated glucuronidation pathway, co-administration of UGT1A1 inducers may alter plasma levels of raltegravir. Raltegravir Potassium 6-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 27704562-3 2016 Since raltegravir is eliminated mainly by metabolism via an UDP-glucuronosyltransferase (UGT) 1 A1-mediated glucuronidation pathway, co-administration of UGT1A1 inducers may alter plasma levels of raltegravir. Raltegravir Potassium 197-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-98 27704562-3 2016 Since raltegravir is eliminated mainly by metabolism via an UDP-glucuronosyltransferase (UGT) 1 A1-mediated glucuronidation pathway, co-administration of UGT1A1 inducers may alter plasma levels of raltegravir. Raltegravir Potassium 197-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 27696440-7 2016 Since raltegravir is eliminated mainly by metabolism via a uridine diphosphate glucuronosyltransferase (UGT) 1A1-mediated glucuronidation pathway, co-administration of UGT1A1 inhibitors may increase the plasma levels of q.d. Raltegravir Potassium 6-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-112 28933122-0 2016 [Hepatotoxicity of emodin based on UGT1A1 enzyme-mediated bilirubin in liver microsomes]. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 27696440-7 2016 Since raltegravir is eliminated mainly by metabolism via a uridine diphosphate glucuronosyltransferase (UGT) 1A1-mediated glucuronidation pathway, co-administration of UGT1A1 inhibitors may increase the plasma levels of q.d. Raltegravir Potassium 6-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 168-174 28933122-1 2016 To study the hepatotoxicity of emodin based on bilirubin metabolism mediated by glucuronidation of UGT1A1 enzyme. Bilirubin 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 30550693-3 2016 Recent pharmacogenetic studies on irinotecan have revealed the impact of UGT1A1 polymorphisms on severe adverse effects. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 27832172-4 2016 Screening assays with 12 human expressed UGT enzymes and chemical-inhibition assays demonstrated that glucuronide formation was almost exclusively catalyzed by UGT1A1, UGT1A6, and UGT1A9. Glucuronides 102-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 27832172-7 2016 In conclusion, UGT1A1, UGT1A6, and UGT1A9 were the chief contributors to the regioselective glucuronidation of diosmetin and chrysoeriol in the liver. diosmetin 111-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 27832172-7 2016 In conclusion, UGT1A1, UGT1A6, and UGT1A9 were the chief contributors to the regioselective glucuronidation of diosmetin and chrysoeriol in the liver. chrysoeriol 125-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 27176228-0 2016 Human UDP-Glucuronosyltransferases 1A1, 1A3, 1A9, 2B4 and 2B7 are Inhibited by Diethylstilbestrol. Diethylstilbestrol 79-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 27176228-4 2016 DES (10 muM) could decrease activities of UGT1A1, 1A3, 1A9, 2B4 and 2B7 in catalysing 4-methylumbelliferone (4-Mu) glucuronidation. Diethylstilbestrol 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 27176228-4 2016 DES (10 muM) could decrease activities of UGT1A1, 1A3, 1A9, 2B4 and 2B7 in catalysing 4-methylumbelliferone (4-Mu) glucuronidation. Hymecromone 86-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 27895797-4 2016 The promoter (-1000 bp) and exon 1 regions of UDP glucuronosyltransferase family 1 member A complex locus (UGT1A1) gene family members UGT1A1, UGT1A7 and UGT1A9 were sequenced, and comprehensive analysis of their genetic polymorphisms was performed to determine the association between inherited genetic variations and irinotecan-induced toxicity. Irinotecan 319-329 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 27895797-6 2016 The results of the present study revealed that UGT1A1*6 and UGT1A7*3 are risk factors for irinotecan-induced severe neutropenia, and UGT1A9*1b is associated with severe diarrhea. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 27899892-5 2016 We found a statistically significant increase (nominal p < 0.05) in the expression of UGT1A1, UGT1A3, UGT1A4, UGT1A5, UGT1A6, UGT2B7, and UGT2B17, as well as glucuronidation activities of serotonin, testosterone, and vorinostat during the first 25 years of life. Serotonin 191-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 27899892-5 2016 We found a statistically significant increase (nominal p < 0.05) in the expression of UGT1A1, UGT1A3, UGT1A4, UGT1A5, UGT1A6, UGT2B7, and UGT2B17, as well as glucuronidation activities of serotonin, testosterone, and vorinostat during the first 25 years of life. Testosterone 202-214 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 27899892-5 2016 We found a statistically significant increase (nominal p < 0.05) in the expression of UGT1A1, UGT1A3, UGT1A4, UGT1A5, UGT1A6, UGT2B7, and UGT2B17, as well as glucuronidation activities of serotonin, testosterone, and vorinostat during the first 25 years of life. Vorinostat 220-230 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 27781231-4 2016 The control cells were treated with 10 mM acetaminophen without genistein to compare with the effects of the combination of acetaminophen and genistein on the expression of UGT1A1, 1A6 and 1A9, Nrf2 and Keap1 mRNAs, as well as the expression of Nrf2 and Keap1 proteins, which were tested by western blotting. Acetaminophen 124-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 27781231-4 2016 The control cells were treated with 10 mM acetaminophen without genistein to compare with the effects of the combination of acetaminophen and genistein on the expression of UGT1A1, 1A6 and 1A9, Nrf2 and Keap1 mRNAs, as well as the expression of Nrf2 and Keap1 proteins, which were tested by western blotting. Genistein 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 27895797-0 2016 UGT1A1*6, UGT1A7*3 and UGT1A9*1b polymorphisms are predictive markers for severe toxicity in patients with metastatic gastrointestinal cancer treated with irinotecan-based regimens. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27895797-4 2016 The promoter (-1000 bp) and exon 1 regions of UDP glucuronosyltransferase family 1 member A complex locus (UGT1A1) gene family members UGT1A1, UGT1A7 and UGT1A9 were sequenced, and comprehensive analysis of their genetic polymorphisms was performed to determine the association between inherited genetic variations and irinotecan-induced toxicity. Irinotecan 319-329 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 30550693-4 2016 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 30550693-4 2016 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 30550693-5 2016 For patients showing homozygosity for UGT1A1*28, *6 or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 30550693-5 2016 For patients showing homozygosity for UGT1A1*28, *6 or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 27695274-7 2016 Both UGT1A1 and UGT2B7 were inhibited by the ethanol and aqueous extracts with IC50 values ranging between 9.59-22.76 mug/mL and 110.71-526.65 mug/Ml, respectively. Ethanol 45-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 27676153-6 2016 Fusidic acid also inhibited UGT1A1-catalyzed beta-estradiol glucuronidation activity in human liver microsomes with an IC50 value of 16 muM. Fusidic Acid 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 27676153-6 2016 Fusidic acid also inhibited UGT1A1-catalyzed beta-estradiol glucuronidation activity in human liver microsomes with an IC50 value of 16 muM. Estradiol 45-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 27676153-9 2016 The magnitudes of in vivo interaction (R values) resulting from the inhibition of OATP1B1, UGT1A1, NTCP, and BSEP transport were ~1.9-2.6, 1.1-1.2, 1.0-1.1, and 1.4-1.7, respectively, which are indicative of some degree of inherent toxicity risk, particularly via inhibition of OATP and BSEP. periodate-oxidized adenosine 5'-triphosphate 82-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 27708529-0 2016 Imatinib-induced hyperbilirubinemia with UGT1A1 (*28) promoter polymorphism: first case series in patients with gastrointestinal stromal tumor. Imatinib Mesylate 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 27708529-2 2016 Severe hepatotoxicity associated with imatinib is rare, and relationship to polymorphism of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) expression and related frequency of hyperbilirubinemia or toxicity are not well known. Imatinib Mesylate 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 27708529-8 2016 This association is also timely due to recent FDA requirement for testing UGT1A1 polymorphism for nilotinib, another TKI. nilotinib 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 27767376-6 2016 Although pharmacogenomic studies for HDIs are scarce, available data indicate that therapy with belinostat (UGT1A1), romidepsin (ABCB1), vorinostat (UGT2B17) or VPA (UGT1A6) could be optimized by upfront genotyping. belinostat 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-58 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-58 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-58 26935006-2 2016 Atazanavir (ATV) inhibits UDP-glucuronosyl-transferase 1A1 (UGT1A1), thus increasing unconjugated bilirubin levels. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 27695274-9 2016 Xanthorrhizol was the better inhibitor of UGT1A1 (IC50 11.30 +- 0.27 muM) as compared to UGT2B7 while curcumene did not show any inhibition. xanthorrhizol 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 27116457-4 2016 RESULTS: After adjustment of the tests by the UGT1A1*28 genotype and correction for multiple testing, the A allele of NR1I2-rs10934498 was associated with a decreased exposition and an increased degradation of SN-38, the active metabolite (p = 0.009 and p = 0.017, respectively). Irinotecan 210-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 27372715-6 2016 UDP-glucuronyltransferase (UGT) initial activity screening showed that UGT1A1, UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7, UGT2B15, and UGT2B17 formed desomorphine glucuronide. desomorphine glucuronide 148-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 27428458-8 2016 Psoralidin showed potent and noncompetitive inhibition against UGT1A1, UGT1A7, CYP1A2 and CYP2C8 with IC50 values of 6.12, 0.38, 1.81, 0.28muM, respectively. psoralidin 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 26833852-3 2016 The results showed that most model flavonoids (from a total of 13) were mainly metabolized into glucuronides in the MDCKII-UGT1A1/MRP2 cells and the glucuronides were rapidly excreted. Flavonoids 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 27746618-10 2016 The mutant, but not wild type or heterozygous genotype of SNPs (rs6742078 and rs887829) in UGT1A1 gene, was associated with significantly higher levels of bilirubin. Bilirubin 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 27746618-11 2016 CONCLUSIONS: Higher incidence of pigment stones in South Indians could be due to raised serum bilirubin levels that may be ascribed to variant in the UGT1A1 gene involved in glucuronidation of free bilirubin. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 27746618-11 2016 CONCLUSIONS: Higher incidence of pigment stones in South Indians could be due to raised serum bilirubin levels that may be ascribed to variant in the UGT1A1 gene involved in glucuronidation of free bilirubin. Bilirubin 198-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 27565905-2 2016 We report a case in which gefitinib- and elrotinib-induced severe hepatotoxicity arose in a patient with the uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) and cytochrome p450 3A5 (CYP3A5) poor metabolizer phenotypes. Gefitinib 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-164 27565905-2 2016 We report a case in which gefitinib- and elrotinib-induced severe hepatotoxicity arose in a patient with the uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) and cytochrome p450 3A5 (CYP3A5) poor metabolizer phenotypes. Gefitinib 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 27565905-2 2016 We report a case in which gefitinib- and elrotinib-induced severe hepatotoxicity arose in a patient with the uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) and cytochrome p450 3A5 (CYP3A5) poor metabolizer phenotypes. elrotinib 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-164 27565905-2 2016 We report a case in which gefitinib- and elrotinib-induced severe hepatotoxicity arose in a patient with the uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) and cytochrome p450 3A5 (CYP3A5) poor metabolizer phenotypes. elrotinib 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 26833852-3 2016 The results showed that most model flavonoids (from a total of 13) were mainly metabolized into glucuronides in the MDCKII-UGT1A1/MRP2 cells and the glucuronides were rapidly excreted. Glucuronides 96-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 26833852-3 2016 The results showed that most model flavonoids (from a total of 13) were mainly metabolized into glucuronides in the MDCKII-UGT1A1/MRP2 cells and the glucuronides were rapidly excreted. Glucuronides 149-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 26833852-6 2016 Maximal velocity of glucuronide production MDCK-MRP2/UGT1A1 cells showed a fairly good correlation (R(2) >0.8) with those derived using UGT1A1 microsomes, but other kinetic parameters (e.g., Km ) did not correlate. Glucuronides 20-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 26833852-6 2016 Maximal velocity of glucuronide production MDCK-MRP2/UGT1A1 cells showed a fairly good correlation (R(2) >0.8) with those derived using UGT1A1 microsomes, but other kinetic parameters (e.g., Km ) did not correlate. Glucuronides 20-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 27533851-0 2016 Polymorphisms in SLCO1B1 and UGT1A1 are associated with sorafenib-induced toxicity. Sorafenib 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 27533851-7 2016 CONCLUSION: Polymorphisms in SLCO1B1 and UGT1A1 are associated with several different sorafenib side effects. Sorafenib 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 28920386-7 2016 In conclusion, the results indicated that chloroform extract showed different inhibitory effects on UGTs and UGT1A1 activity, which may be one of the mechanisms of liver injury induced by Genkwa Flos. Chloroform 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 28920386-0 2016 [In vitro effects of Genkwa Flos chloroform extract on activity of human liver microsomes UGTs and UGT1A1]. Chloroform 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Nevirapine 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 28920386-2 2016 In the present study, 4-nitrophenol(4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC. 4-nitrophenol 22-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 28920386-2 2016 In the present study, 4-nitrophenol(4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC. 4-nonylphenol 36-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 28920386-2 2016 In the present study, 4-nitrophenol(4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC. Estradiol 46-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 27529203-3 2016 We also predicted potential dietary supplement-drug interactions for beta-cryptoxanthin via UGT1A1 inhibition. Beta-Cryptoxanthin 69-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 27529203-5 2016 beta-Cryptoxanthin mildly inhibited UGT1A1, UGT1A3, and UGT1A4, with IC50 values of 18.8 +- 2.07, 28.3 +- 4.40 and 34.9 +- 5.98 muM, respectively. Beta-Cryptoxanthin 0-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 27529203-6 2016 Canthaxanthin weakly inhibited UGT1A1 and UGT1A3, with IC50 values of 38.5 +- 4.65 and 41.2 +- 3.14 muM, respectively; and lutein inhibited UGT1A1 and UGT1A4, with IC50 values of 45.5 +- 4.01 and 28.7 +- 3.79 muM, respectively. Canthaxanthin 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 27529203-6 2016 Canthaxanthin weakly inhibited UGT1A1 and UGT1A3, with IC50 values of 38.5 +- 4.65 and 41.2 +- 3.14 muM, respectively; and lutein inhibited UGT1A1 and UGT1A4, with IC50 values of 45.5 +- 4.01 and 28.7 +- 3.79 muM, respectively. Canthaxanthin 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 27529203-7 2016 Among the tested xanthophyll-UGT pairs, beta-cryptoxanthin showed the strongest competitive inhibition of UGT1A1 (Ki, 12.2 +- 0.985 muM). Beta-Cryptoxanthin 40-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 27529203-8 2016 In addition, we predicted the risk of UGT1A1 inhibition in vivo using the reported maximum plasma concentration after oral administration of beta-cryptoxanthin in humans. Beta-Cryptoxanthin 141-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 26626330-10 2016 Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Ritonavir 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 248-254 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)-imidazo(2,1-b)thiazole 70-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 248-254 27325020-6 2016 However, a PGG glucuronide was absent in the UGT1A1 system. Glucuronides 15-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 26829336-11 2016 Resveratrol was a competitive inhibitor of UGT1A1, UGT1A9 and HLM; quercetin and kaempferol were inhibitors of all transferases under investigation except UGT2B15. Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 27380166-0 2016 Erratum to: UDP-glucuronosyltransferase 1A1*6 and *28 polymorphisms as indicators of initial dose level of irinotecan to reduce risk of neutropenia in patients receiving FOLFIRI for colorectal cancer. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 26710796-0 2016 UDP-glucuronosyltransferase 1A1*6 and *28 polymorphisms as indicators of initial dose level of irinotecan to reduce risk of neutropenia in patients receiving FOLFIRI for colorectal cancer. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 26632033-10 2016 ABT-751 is metabolized by UGT1A8 and to a lesser extent UGT1A4 and UGT1A1. ABT751 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 27057738-2 2016 We evaluated the role of variations in the bilirubin uridine-5-diphosphate (UDP)-glucuronosyltransferase gene (UGT1A1) in unconjugated hyperbilirubinemia development during chemotherapy in pediatric patients with leukemia. Bilirubin 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27057738-2 2016 We evaluated the role of variations in the bilirubin uridine-5-diphosphate (UDP)-glucuronosyltransferase gene (UGT1A1) in unconjugated hyperbilirubinemia development during chemotherapy in pediatric patients with leukemia. Uridine Diphosphate 53-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 27057738-3 2016 METHODS: UGT1A1 allelic variations were evaluated in 25 Japanese pediatric leukemia patients with hyperbilirubinemia (peak serum bilirubin concentration 3.57 +- 1.02 mg/dl) and 25 control patients without hyperbilirubinemia (0.92 +- 0.32 mg/dl) by PCR-direct sequencing. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 27220761-0 2016 UGT1A1 gene polymorphism is associated with toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26739304-0 2016 High Resectability Rate of Initially Unresectable Colorectal Liver Metastases After UGT1A1-Adapted High-Dose Irinotecan Combined with LV5FU2 and Cetuximab: A Multicenter Phase II Study (ERBIFORT). Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 26739304-14 2016 It seemed to be well-tolerated among healthy selected patients thanks to irinotecan dose adaptation according to UGT1A1 pharmacogenomics status. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 27220761-1 2016 OBJECTIVES: To investigate the relationship between uridine diphosphate glucoronosyltransferase 1A1 (UGT1A1)*28/*6 and toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer (CRC) in Xinjiang Uygur and Han population. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-99 27220761-1 2016 OBJECTIVES: To investigate the relationship between uridine diphosphate glucoronosyltransferase 1A1 (UGT1A1)*28/*6 and toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer (CRC) in Xinjiang Uygur and Han population. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 288-298 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 27220761-11 2016 CONCLUSION: UGT1A1 gene polymorphism predicts irinotecan-related adverse reactions in advanced CRC patients of Xinjiang Uygur and Han nationality; UGT1A1 gene polymorphism is correlated with efficacy and prognosis in Uygur nationality, but only related to prognosis in Han nationality in irinotecan-based chemotherapy. Irinotecan 288-298 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 27160286-1 2016 PURPOSE: UGT1A1*28/*6 as predictors of severe irinotecan-related diarrhea (SIRD) were duplicated by many studies. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 27029846-8 2016 Concomitant administration of non-ritonavir-boosted atazanavir decreased elvitegravir clearance by 35%, likely due to UDP-glucuronosyl transferase (UGT) 1A1 inhibition. Ritonavir 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-156 27029846-8 2016 Concomitant administration of non-ritonavir-boosted atazanavir decreased elvitegravir clearance by 35%, likely due to UDP-glucuronosyl transferase (UGT) 1A1 inhibition. Atazanavir Sulfate 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-156 27160286-0 2016 A novel genetic score model of UGT1A1 and TGFB pathway as predictor of severe irinotecan-related diarrhea in metastatic colorectal cancer patients. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 26898568-1 2016 PURPOSE: Dolutegravir (DTG) is primarily metabolized by UGT1A1 with CYP3A as a minor route. dolutegravir 9-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 27089846-4 2016 The results demonstrated that both tolcapone and entacapone exhibited inhibitory effects on UGT1A1, UGT1A7, UGT1A9 and UGT1A10. Tolcapone 35-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 27089846-4 2016 The results demonstrated that both tolcapone and entacapone exhibited inhibitory effects on UGT1A1, UGT1A7, UGT1A9 and UGT1A10. entacapone 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 26862009-0 2016 Impact of UGT1A1 genotype upon toxicities of combination with low-dose irinotecan plus platinum. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 26862009-0 2016 Impact of UGT1A1 genotype upon toxicities of combination with low-dose irinotecan plus platinum. Platinum 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 26862009-1 2016 AIM: Irinotecan-induced severe toxicities are possibly related to UGT1A1*6 and *28 genotypes. Irinotecan 5-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 26862009-2 2016 However, the correlation between UGT1A1 polymorphisms and the risk of toxicities induced by low-dose irinotecan plus platinum combination therapy still remains controversial. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26862009-2 2016 However, the correlation between UGT1A1 polymorphisms and the risk of toxicities induced by low-dose irinotecan plus platinum combination therapy still remains controversial. Platinum 117-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26862009-3 2016 This prospective observational study aimed to examine the correlation between UGT1A1 genotypes and clinical outcomes of low-dose irinotecan (median 60 mg/m(2) , range 25-115 mg/m(2) ) plus platinum in Japanese patients with solid tumors. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 26862009-10 2016 CONCLUSION: UGT1A1 genotype has a major impact on the increased risk of severe hematological toxicities, even in low-dose irinotecan regimens. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 26862009-11 2016 UGT1A1 genotypes are useful biomarkers for predicting severe hematological toxicities in patients treated with irinotecan plus platinum analog. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26862009-11 2016 UGT1A1 genotypes are useful biomarkers for predicting severe hematological toxicities in patients treated with irinotecan plus platinum analog. Platinum 127-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 27385990-0 2016 Prospective study of the UGT1A1*27 gene polymorphism during irinotecan therapy in patients with lung cancer: Results of Lung Oncology Group in Kyusyu (LOGIK1004B). Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 27385990-2 2016 To evaluate the effect of the UGT1A1*27 polymorphism in irinotecan therapy, we conducted a prospective study. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 27385990-10 2016 In an evaluation of the side effects of irinotecan, patients with UGT1A1*28 and UGT1A1*6 polymorphisms had a higher tendency to experience febrile neutropenia than wild type (25% and 32% vs. 14%). Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 27385990-10 2016 In an evaluation of the side effects of irinotecan, patients with UGT1A1*28 and UGT1A1*6 polymorphisms had a higher tendency to experience febrile neutropenia than wild type (25% and 32% vs. 14%). Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 27089846-5 2016 In contrast to entacapone, tolcapone exhibited more potent inhibitory effects on UGT1A1, UGT1A7, and UGT1A10, while their inhibitory potentials against UGT1A9 were comparable. Tolcapone 27-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 27089846-6 2016 It is noteworthy that the inhibition constants (Ki) of tolcapone and entacapone against bilirubin-O-glucuronidation in human liver microsomes (HLM) are determined as 0.68muM and 30.82muM, respectively, which means that the inhibition potency of tolcapone on UGT1A1 mediated bilirubin-O-glucuronidation in HLM is much higher than that of entacapone. Tolcapone 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 258-264 27089846-6 2016 It is noteworthy that the inhibition constants (Ki) of tolcapone and entacapone against bilirubin-O-glucuronidation in human liver microsomes (HLM) are determined as 0.68muM and 30.82muM, respectively, which means that the inhibition potency of tolcapone on UGT1A1 mediated bilirubin-O-glucuronidation in HLM is much higher than that of entacapone. entacapone 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 258-264 27089846-7 2016 Furthermore, the potential risks of tolcapone or entacapone via inhibition of human UGT1A1 were quantitatively predicted by the ratio of the areas under the plasma drug concentration-time curve (AUC). Tolcapone 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 27089846-7 2016 Furthermore, the potential risks of tolcapone or entacapone via inhibition of human UGT1A1 were quantitatively predicted by the ratio of the areas under the plasma drug concentration-time curve (AUC). entacapone 49-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 27089846-8 2016 The results indicate that tolcapone may result in significant increase in AUC of bilirubin or the drugs primarily metabolized by UGT1A1, while entacapone is unlikely to cause a significant DDI through inhibition of UGT1A1. Tolcapone 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 27296832-1 2016 BACKGROUND: To investigate the associations between atorvastatin-induced liver injury (AILI) and polymorphisms in eight genes possibly involved in the hepatic metabolism (CYP2C9, CYP2C19, CYP3A4, CYP3A5 and UGT1A1) and membrane transport (ABCB1, ABCG2 and SLCO1B1) of atorvastatin, we genotyped 30 AILI and 414 non-AILI patients recruited at BioBank Japan for 15 single nucleotide polymorphisms (SNPs). Atorvastatin 52-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 207-213 26898568-1 2016 PURPOSE: Dolutegravir (DTG) is primarily metabolized by UGT1A1 with CYP3A as a minor route. dolutegravir 23-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 26758854-0 2016 Efflux Transport Characterization of Resveratrol Glucuronides in UDP-Glucuronosyltransferase 1A1 Transfected HeLa Cells: Application of a Cellular Pharmacokinetic Model to Decipher the Contribution of Multidrug Resistance-Associated Protein 4. resveratrol glucuronides 37-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-96 27323053-7 2016 The serum bilirubin levels seem to be affected by the homozygosity or heterozygosity of the UGT1A1 gene mutation; 211G>A homozygous mutation is an important factor that causes a rise in bilirubin in neonates with unconjugated hyperbilirubinemia. Bilirubin 10-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 27323053-7 2016 The serum bilirubin levels seem to be affected by the homozygosity or heterozygosity of the UGT1A1 gene mutation; 211G>A homozygous mutation is an important factor that causes a rise in bilirubin in neonates with unconjugated hyperbilirubinemia. Bilirubin 189-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 29874021-9 2016 The sciadopitysin competitively inhibited the formation of 4-MU-O-glucuronide by UGT1A1, UGT1A3, UGT1A8, and UGT1A10. 4-mu-o-glucuronide 59-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 29874021-10 2016 At the same time, the inhibition of NCHN-O-glucuronidation by UGT1A1 was consistent with the competitive inhibition. nchn-o 36-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 27099654-7 2016 Phenobarbitone being an inducer of enzyme UDPGT is used as the first line of treatment and is not teratogenic in the low doses used. Phenobarbital 0-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-47 27122675-1 2016 AIM: To examine the predictive effects of baseline serum bilirubin levels and UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism on response of colorectal cancer to irinotecan-based chemotherapy. Irinotecan 168-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-115 27122675-9 2016 RESULTS: Among the 120 mCRC patients, the serum bilirubin level was significantly different between the UGT1A1*28 wild-type and mutant genotypes. Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 27122675-16 2016 CONCLUSION: CoBil and UGT1A1*28 are both independent biomarkers for predicting the treatment response of mCRC patients to irinotecan-based chemotherapy. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 26417955-1 2016 The antiretroviral protease inhibitor atazanavir inhibits hepatic uridine diphosphate glucuronosyltransferase (UGT) 1A1, thereby preventing the glucuronidation and elimination of bilirubin. Atazanavir Sulfate 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-119 26417955-1 2016 The antiretroviral protease inhibitor atazanavir inhibits hepatic uridine diphosphate glucuronosyltransferase (UGT) 1A1, thereby preventing the glucuronidation and elimination of bilirubin. Bilirubin 179-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-119 26417955-3 2016 Risk for bilirubin-related discontinuation is highest among individuals who carry two UGT1A1 decreased function alleles (UGT1A1*28 or *37). Bilirubin 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 26417955-3 2016 Risk for bilirubin-related discontinuation is highest among individuals who carry two UGT1A1 decreased function alleles (UGT1A1*28 or *37). Bilirubin 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 26417955-4 2016 We summarize published literature that supports this association and provide recommendations for atazanavir prescribing when UGT1A1 genotype is known (updates at www.pharmgkb.org). Atazanavir Sulfate 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 26758854-3 2016 Here, we aimed to characterize the efflux transport of resveratrol glucuronides using UGT1A1-overexpressing HeLa cells (HeLa1A1 cells), and to determine the contribution of multidrug resistance-associated protein (MRP) 4 to cellular excretion of the glucuronides. resveratrol glucuronides 55-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 26758854-3 2016 Here, we aimed to characterize the efflux transport of resveratrol glucuronides using UGT1A1-overexpressing HeLa cells (HeLa1A1 cells), and to determine the contribution of multidrug resistance-associated protein (MRP) 4 to cellular excretion of the glucuronides. Glucuronides 67-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 27080842-6 2016 Other tests are also commonly performed to address the toxicity of certain anticancer drugs (DPYD-capecitabine, UGT1A1-irinotecan, TPMT 6-mercaptopurine). Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 27461651-2 2016 DPYD*2A/*5A/*9A and UGT1A1 * 6/*28 polymorphisms are related to the toxicity of fluorouracil drugs and irinotecan, respectively. Fluorouracil 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 27461651-2 2016 DPYD*2A/*5A/*9A and UGT1A1 * 6/*28 polymorphisms are related to the toxicity of fluorouracil drugs and irinotecan, respectively. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 26313268-2 2016 Polymorphisms that reduce UGT1A1 function could result in increased belinostat exposure and toxicities. belinostat 68-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 26313268-10 2016 UGT1A1 polymorphisms were associated with increased systemic belinostat exposure, increased AcK, and increased incidence of toxicities, particularly at doses > 400 mg/m(2) /24 h. belinostat 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26637161-2 2016 Belinostat is predominantly metabolized by UGT1A1, which is polymorphic. belinostat 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 26637161-6 2016 This model and subsequent simulations supported additional PK/toxicity and pharmacogenomics/toxicity analyses to suggest a UGT1A1 genotype-based dose adjustment to normalize belinostat exposure and allow for more tolerable therapy. belinostat 174-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 27034809-6 2016 The presence of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *28 polymorphism is associated with toxicity from irinotecan, although it has not been universally applied. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-67 27034809-6 2016 The presence of the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) *28 polymorphism is associated with toxicity from irinotecan, although it has not been universally applied. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26189431-3 2016 The in vitro studies using atazanavir as a selective inhibitor of UGT1A1 and -1A3 indicated that these enzymes contribute ~55% toward the overall glucuronidation pathway. Atazanavir Sulfate 27-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-81 27025983-6 2016 Dimerization changed the chemical regioselectivity, substrate-binding affinity, and enzymatic activity of UGT1A1 and UGT1A9 in glucuronidation of quercetin. Quercetin 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 27025983-7 2016 These findings provide molecular insights into the consequences of homozygous and heterozygous UGT1A1 and UGT1A9 allozymes expression on quercetin glucuronidation. Quercetin 137-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 26781906-0 2016 Three-dimensional polyacrylamide gel-based DNA microarray method effectively identifies UDP-glucuronosyltransferase 1A1 gene polymorphisms for the correct diagnosis of Gilbert"s syndrome. polyacrylamide 18-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-119 26781906-2 2016 The T-3279G mutation in the phenobarbital responsive enhancer module (PBREM), the TA-insertion in the TATA box, creating the A(TA)7TAA motif instead of A(TA)6TAA and the G211A mutation in coding exon 1, particularly in Asian populations, of the human UGT1A1 gene are the three common genotypes found in patients with Gilbert"s syndrome. Phenobarbital 28-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 26781906-4 2016 In this study, to the best of our knowledge, we established a three-dimensional polyacrylamide gel-based DNA microarray method for the first time, in order to study UGT1A1 gene polymorphisms. polyacrylamide 80-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 26781906-7 2016 The data from the current study demonstrate that the three-dimensional polyacrylamide gel microarray method has the potential to be applied as a useful, reliable and cost-effective tool to detect the T-3279G, the A(TA)6/7TAA and the G211A mutations of the UGT1A1 gene in patients with hyperbilirubinemia and thereby aid in the diagnosis of Gilbert"s syndrome. polyacrylamide 71-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 256-262 26146896-2 2016 Indeed, this is the first report that interested in the study of polymorphisms in genes encoded for enzymes involved in the bilirubin metabolism: rs 4149056 of SLCO1B1 and rs4149000 of SLCO1A2 in combination with rs8175347 and rs887829 of UGT1A1 in order to find a correlation between the polymorphisms studied and the presence of gallstones in a population of sickle cell anemia (SCA) pediatric Tunisians. Bilirubin 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 239-245 26857783-4 2016 RESULTS AND CONCLUSION: UGT1A1*6 and UGT1A1*28 polymorphisms were associated with severe neutropenia (p = 0.025, p = 0.022, respectively) but not diarrhea (p = 0.343, p = 0.185, respectively), and ABCB1- 3435C>T polymorphism was not associated with irinotecan induced severe toxicities (p = 0.457, p = 0.161, respectively). Irinotecan 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26857783-4 2016 RESULTS AND CONCLUSION: UGT1A1*6 and UGT1A1*28 polymorphisms were associated with severe neutropenia (p = 0.025, p = 0.022, respectively) but not diarrhea (p = 0.343, p = 0.185, respectively), and ABCB1- 3435C>T polymorphism was not associated with irinotecan induced severe toxicities (p = 0.457, p = 0.161, respectively). Irinotecan 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 26189431-4 2016 Therefore, a clinical study was conducted to assess the potential for drug interaction between pradigastat and probenecid (purported general UGT inhibitor) or atazanavir (selective UGT1A1, -1A3 inhibitor). pradigastat 95-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 26502886-8 2016 In the presence of 50 nM curcumin, the clearance of resveratrol-3-O-glucuronide and resveratrol-3-O-sulfate reduced in both MRP2-overexpressing MDCKII-UGT1A1 cells and Human UGT1A9-overexpressing HeLa cells. Curcumin 25-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 26502886-8 2016 In the presence of 50 nM curcumin, the clearance of resveratrol-3-O-glucuronide and resveratrol-3-O-sulfate reduced in both MRP2-overexpressing MDCKII-UGT1A1 cells and Human UGT1A9-overexpressing HeLa cells. resveratrol-4'-O-glucuronide 52-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-85 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 26773202-1 2016 Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. Irinotecan 271-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 26773202-3 2016 Belinostat, approved for the treatment of peripheral T-cell lymphoma, is primarily glucuronidated by UGT1A1. belinostat 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 26502886-8 2016 In the presence of 50 nM curcumin, the clearance of resveratrol-3-O-glucuronide and resveratrol-3-O-sulfate reduced in both MRP2-overexpressing MDCKII-UGT1A1 cells and Human UGT1A9-overexpressing HeLa cells. Resveratrol-3-O-sulfate 84-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 26773202-4 2016 Recent preclinical and clinical data showed that UGT1A1*28 was associated with reduced glucuronidation in human liver microsomes, while in a retrospective analysis of a Phase I trial with patients receiving belinostat UGT1A1*60 was predominantly associated with increased belinostat plasma concentrations. belinostat 207-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 26773202-4 2016 Recent preclinical and clinical data showed that UGT1A1*28 was associated with reduced glucuronidation in human liver microsomes, while in a retrospective analysis of a Phase I trial with patients receiving belinostat UGT1A1*60 was predominantly associated with increased belinostat plasma concentrations. belinostat 207-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 218-224 26773202-4 2016 Recent preclinical and clinical data showed that UGT1A1*28 was associated with reduced glucuronidation in human liver microsomes, while in a retrospective analysis of a Phase I trial with patients receiving belinostat UGT1A1*60 was predominantly associated with increased belinostat plasma concentrations. belinostat 272-282 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 26830078-0 2016 Correlation of UGT1A1(*)28 and (*)6 polymorphisms with irinotecan-induced neutropenia in Thai colorectal cancer patients. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26547877-5 2016 DPhP and DNOP weakly inhibited the activities of UGT1A1, UGT1A7, and UGT1A8. dphp 0-4 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 26547877-5 2016 DPhP and DNOP weakly inhibited the activities of UGT1A1, UGT1A7, and UGT1A8. di-n-octyl phthalate 9-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 26830078-1 2016 UDP-glucuronosyltransferase1A1 (UGT1A1) polymorphisms have been related with irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 26830078-1 2016 UDP-glucuronosyltransferase1A1 (UGT1A1) polymorphisms have been related with irinotecan toxicity. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 26830078-2 2016 The purpose of this study was to determine the associations between UGT1A1(*)28 and (*)6 polymorphisms and irinotecan toxicity in Thai patients with metastatic colorectal cancer. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 26830078-10 2016 Both UGT1A1(*)28 and (*)6 polymorphisms may have an increased risk of irinotecan-induced neutropenia in Thai colorectal cancer patients. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 28868871-2 2016 In the present study, 4-nitrophenol (4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC, respectively. 4-nitrophenol 22-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 25869015-6 2016 For SN-38 AUC0-24 and the glucuronidation ratio, we replicated UGT1A1*28 and UGT1A1*93. Irinotecan 4-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 25869015-6 2016 For SN-38 AUC0-24 and the glucuronidation ratio, we replicated UGT1A1*28 and UGT1A1*93. Irinotecan 4-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 25869015-7 2016 In addition to UGT1A1*28, this study independently validated UGT1A1*93 and SLCO1B1*1b as new predictors of irinotecan neutropenia. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 28868871-2 2016 In the present study, 4-nitrophenol (4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC, respectively. 4-nitrophenol 37-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 28868871-2 2016 In the present study, 4-nitrophenol (4-NP) and beta-estradiol were elected as substrates to determine activities of UGTs and UGT1A1 by UV and HPLC, respectively. Estradiol 47-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 26808671-0 2016 Phenotyping of UGT1A1 Activity Using Raltegravir Predicts Pharmacokinetics and Toxicity of Irinotecan in FOLFIRI. Raltegravir Potassium 37-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26820647-0 2016 Molecular Genetics External Quality Assessment Pilot Scheme for Irinotecan-Related UGT1A1 Genotyping in China. Irinotecan 64-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 26820647-2 2016 Molecular testing for UGT1A1 genotyping is increasingly required in China for optimum irinotecan administration. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 26808671-0 2016 Phenotyping of UGT1A1 Activity Using Raltegravir Predicts Pharmacokinetics and Toxicity of Irinotecan in FOLFIRI. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26811156-0 2016 Prospective analysis of UGT1A1 promoter polymorphism for irinotecan dose escalation in metastatic colorectal cancer patients treated with bevacizumab plus FOLFIRI as the first-line setting: study protocol for a randomized controlled trial. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26808671-1 2016 BACKGROUND: Irinotecan toxicity correlates with UGT1A1 activity. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26811156-0 2016 Prospective analysis of UGT1A1 promoter polymorphism for irinotecan dose escalation in metastatic colorectal cancer patients treated with bevacizumab plus FOLFIRI as the first-line setting: study protocol for a randomized controlled trial. FOLFIRI regimen 155-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26098842-2 2016 This study evaluates the cost-effectiveness of UGT1A1 genotyping in patients with metastatic colorectal cancer undergoing irinotecan-based chemotherapy compared to no testing from the perspective of the German statutory health insurance. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 26811156-3 2016 Therefore, we plan to explore the effect of the genetic polymorphism of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) for irinotecan detoxification in mCRC patients. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-119 26811156-3 2016 Therefore, we plan to explore the effect of the genetic polymorphism of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) for irinotecan detoxification in mCRC patients. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 26811156-6 2016 METHODS/DESIGN: This trial is a prospective, multicenter, randomized clinical trial comparing UGT1A1 promoter polymorphism for irinotecan dose escalation in mCRC patients administered with bevacizumab plus FOLFIRI as the first-line setting. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 26811156-8 2016 The study group receive a biweekly FOLFIRI regimen, with irinotecan dose escalation based on UGT1A1 genotyping; whereas the control group receive the conventional biweekly FOLFIRI regimen without UGT1A1 genotyping. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 26811156-10 2016 DISCUSSION: Patients with mCRC undergoing UGT1A1 genotyping may receive escalated doses of irinotecan for a potentially more favorable clinical response and outcome, in addition to comparable toxicities. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 26872008-0 2016 Phase II and UGT1A1 Polymorphism Study of Two Different Irinotecan Dosages Combined with Cisplatin as First-Line Therapy for Advanced Gastric Cancer. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 27725437-2 2016 Trovafloxacin is mainly metabolized to its acyl-glucuronide by uridine 5"-diphosphate (UDP)-glucuronosyltransferase (UGT) 1A1. trovafloxacin 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-125 27725437-2 2016 Trovafloxacin is mainly metabolized to its acyl-glucuronide by uridine 5"-diphosphate (UDP)-glucuronosyltransferase (UGT) 1A1. acyl-glucuronide 43-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-125 27725437-2 2016 Trovafloxacin is mainly metabolized to its acyl-glucuronide by uridine 5"-diphosphate (UDP)-glucuronosyltransferase (UGT) 1A1. Uridine Diphosphate 63-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-125 27725437-4 2016 A UGT1A1-induced cell model was developed and the toxicity of trovafloxacin acyl-glucuronide was evaluated. trovafloxacin acyl-glucuronide 62-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 27725437-5 2016 The UGT1A1-induced cell model was developed by treating HepG2 cells with chrysin for 48 h. Chemokine (C-X-C motif) ligand 2, a cytokine involved in drug-induced liver injury, was uniquely induced by trovafloxacin in the UGT1A1-induced HepG2 cells. trovafloxacin 199-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 27725437-7 2016 An in vivo animal study further demonstrated the importance of UGT1A1 in the trovafloxacin-induced liver toxicity. trovafloxacin 77-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 26977326-0 2016 Pegvisomant-Induced Cholestatic Hepatitis in an Acromegalic Patient with UGT1A1 (*) 28 Mutation. pegvisomant 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 25545261-1 2016 BACKGROUND: UGT1A1 is a polymorphic enzyme that has been associated with irinotecan drug metabolisms. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 26512042-9 2016 Axitinib N-glucuronidation was primarily catalyzed by UDP-glucuronosyltransferase (UGT) UGT1A1, which was verified by chemical inhibitors and UGT1A1 null expressers, with lesser contributions from UGTs 1A3, 1A9, and 1A4. Axitinib 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 26512042-9 2016 Axitinib N-glucuronidation was primarily catalyzed by UDP-glucuronosyltransferase (UGT) UGT1A1, which was verified by chemical inhibitors and UGT1A1 null expressers, with lesser contributions from UGTs 1A3, 1A9, and 1A4. Axitinib 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 27072236-14 2016 Furthermore, as 5.FU and irinotecan metabolism is complex, numerous genes in addition to DPD and UGT1A1 should be investigated. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 26666247-3 2016 The Gilbert genotype, the UGT1A1*28 allele, is the major known genetic cause of variation in bilirubin. Bilirubin 93-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 26666247-8 2016 RESULTS: Per UGT1A1*28 allele, odds were increased for any need of supplementary oxygen (odds ratio 1.26; 1.05-1.50) and for BPD (odds ratio 1.71; 1.23-2.39), the need of supplementary oxygen increased by 6.38 days (1.87-10.89), and chance per day of no longer needing supplementary oxygen was reduced (hazard rate 0.84; 0.76-0.93). Oxygen 81-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 26666247-8 2016 RESULTS: Per UGT1A1*28 allele, odds were increased for any need of supplementary oxygen (odds ratio 1.26; 1.05-1.50) and for BPD (odds ratio 1.71; 1.23-2.39), the need of supplementary oxygen increased by 6.38 days (1.87-10.89), and chance per day of no longer needing supplementary oxygen was reduced (hazard rate 0.84; 0.76-0.93). Oxygen 185-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 26666247-8 2016 RESULTS: Per UGT1A1*28 allele, odds were increased for any need of supplementary oxygen (odds ratio 1.26; 1.05-1.50) and for BPD (odds ratio 1.71; 1.23-2.39), the need of supplementary oxygen increased by 6.38 days (1.87-10.89), and chance per day of no longer needing supplementary oxygen was reduced (hazard rate 0.84; 0.76-0.93). Oxygen 185-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 26666247-13 2016 CONCLUSIONS: Compared to the common genotype, UGT1A1*28 genotypes were associated with an increased need of oxygen supplementation and risk of BPD in very preterm newborns. Oxygen 108-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 26068521-0 2016 Metabolism of the anthelmintic drug niclosamide by cytochrome P450 enzymes and UDP-glucuronosyltransferases: metabolite elucidation and main contributions from CYP1A2 and UGT1A1. Niclosamide 36-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 26068521-11 2016 Although seven UGT enzymes were able to catalyze glucuronidation of niclosamide, UGT1A1 and 1A3 were the enzymes showed the highest metabolic activities. Niclosamide 68-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-95 26247833-0 2016 Raloxifene glucuronidation in liver and intestinal microsomes of humans and monkeys: contribution of UGT1A1, UGT1A8 and UGT1A9. Raloxifene Hydrochloride 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 26134155-13 2016 In vitro phenotyping experiments demonstrated that cabotegravir was metabolized by UDP-glucuronosyltransferase (UGT) 1A1 and UGT1A9. cabotegravir 51-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-120 26068521-12 2016 Activity correlation analyses demonstrated that UGT1A1 played a predominant role in hepatic glucuronidation of niclosamide, whereas the role of UGT1A3 was negligible. Niclosamide 111-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26247833-3 2016 In this study, the in vitro glucuronidation of raloxifene in humans and monkeys was examined using liver and intestinal microsomes and recombinant UGT enzymes (UGT1A1, UGT1A8 and UGT1A9). Raloxifene Hydrochloride 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 26068521-14 2016 In conclusion, niclosamide was subjected to efficient metabolic reactions hydroxylation and glucuronidation, wherein CYP1A2 and UGT1A1 were the main contributing enzymes, respectively. Niclosamide 15-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 27803477-5 2016 1) In 2005, information regarding the genetic polymorphism of UGT1A1*28 was described in the package insert of the drug irinotecan in the United States. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 27803477-7 2016 Through clinical research, we demonstrated that UGT1A1*6 was a significant factor for neutropenia, as induced by irinotecan. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26642944-4 2015 Lapatinib exhibited potent competitive inhibition against UGT1A1 activity with a Ki of 0.5 muM. Lapatinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 26642944-6 2015 The TKIs also exerted intermediate inhibition against several UGTs (i.e., UGT1A7 by lapatinib; UGT1A1 by imatinib; UGT1A4, 1A7 and 1A9 by axitinib; and UGT1A9 by vandetanib). Imatinib Mesylate 105-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 26642944-7 2015 Results from modeling for the quantitative prediction of DDI risk indicated that the coadministration of lapatinib or imatinib at clinical doses could result in a significant increase in AUC of drugs primarily cleared by UGT1A1 or 2B17. Lapatinib 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 221-227 26642944-7 2015 Results from modeling for the quantitative prediction of DDI risk indicated that the coadministration of lapatinib or imatinib at clinical doses could result in a significant increase in AUC of drugs primarily cleared by UGT1A1 or 2B17. Imatinib Mesylate 118-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 221-227 26642944-8 2015 Lapatinib and imatinib may cause clinically significant DDIs when co-administered UGT1A1 or 2B17 substrates. Lapatinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 26642944-8 2015 Lapatinib and imatinib may cause clinically significant DDIs when co-administered UGT1A1 or 2B17 substrates. Imatinib Mesylate 14-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 26628212-0 2015 Serum bilirubin concentration is modified by UGT1A1 haplotypes and influences risk of type-2 diabetes in the Norfolk Island genetic isolate. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 26628212-5 2015 Strong linkage disequilibrium was observed across a 200 kb region spanning the UDP-glucuronosyltransferase family, including UGT1A1, an enzyme known to metabolise bilirubin. Bilirubin 163-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 26628212-9 2015 CONCLUSIONS: In summary, a pedigree-based GWAS of blood-based clinical traits in the Norfolk Island population has identified variants within the UDPGT family directly associated with serum bilirubin levels, which is in turn implicated with reduced risk of developing type-2 diabetes within this population. Bilirubin 190-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-151 25778466-0 2015 A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-12 26664141-0 2015 The relationship between UGT1A1 gene polymorphism and irinotecan effect on extensive-stage small-cell lung cancer. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 26664141-11 2015 CONCLUSION: For irinotecan-based regimens in E-SCLC, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for severe adverse events. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 26664141-11 2015 CONCLUSION: For irinotecan-based regimens in E-SCLC, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for severe adverse events. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 26223945-5 2015 RESULTS: One UGT1A1 SNP of rs887829 (C>T) exhibited significant association with stable warfarin doses in the study population and subgroup. Warfarin 91-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 26223945-10 2015 CONCLUSION: Our results suggest that UGT1A1 could be a determinant of stable warfarin doses. Warfarin 77-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 117-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 25216634-2 2015 Irinotecan-induced diarrhea is closely related with the UDP-glucuronosyltransferase 1A1-catalyzed glucuronidation of SN-38 which has been widely regarded to be the toxic substance basis of irinotecan. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 26220753-1 2015 Crigler-Najjar syndrome presents as severe unconjugated hyperbilirubinemia and is characteristically caused by a mutation in the UGT1A1 gene, encoding the enzyme responsible for bilirubin glucuronidation. Bilirubin 61-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 26407805-7 2015 UGT1A1, 1A3, 1A7, 1A8, 1A9, 1A10, 2B4, 2B7 and 2B17 participated in the formation of 4"-O-G, while UGT2B17 demonstrated the highest catalytic activity in this biotransformation. 4"-o-g 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26494856-0 2015 High-Dose FOLFIRI plus Bevacizumab in the Treatment of Metastatic Colorectal Cancer Patients with Two Different UGT1A1 Genotypes: FFCD 0504 Study. FOLFIRI regimen 10-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 26494856-2 2015 UDP-glucuronosyltransferase: (UGT1A1) polymorphism may be predictive of toxicity and efficacy of irinotecan. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 26494856-3 2015 This phase II study aimed to evaluate the combination of high-dose FOLFIRI plus bevacizumab in patients with previously untreated metastatic colorectal cancer (MCRC) based on their UGT1A1 genotype. FOLFIRI regimen 67-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-90 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 160-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-90 25778466-1 2015 The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Irinotecan 160-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 26692528-10 2015 CONCLUSIONS: Our study showed that mCRC patients with UGT1A1 *1/*1 and *1/*28 genotypes could receive escalated doses of irinotecan to obtain a more favorable clinical outcome without significant AEs. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 26543529-0 2015 Serum Total Bilirubin, not Cholelithiasis, is Influenced by UGT1A1 Polymorphism, Alpha Thalassemia and beta(s) Haplotype: First Report on Comparison between Arab-Indian and African beta(s) Genes. Bilirubin 12-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 26348140-3 2015 This study aimed to investigate the inhibitory effects of FP extract and its major constituents against human UDP-glucuronosyltransferase 1A1 (UGT1A1), the key enzyme responsible for metabolic elimination of bilirubin. Bilirubin 208-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-141 26348140-3 2015 This study aimed to investigate the inhibitory effects of FP extract and its major constituents against human UDP-glucuronosyltransferase 1A1 (UGT1A1), the key enzyme responsible for metabolic elimination of bilirubin. Bilirubin 208-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 26348140-4 2015 To this end, N-(3-carboxy propyl)-4-hydroxy-1,8-naphthalimide (NCHN), a newly developed specific fluorescent probe for UGT1A1, was used to evaluate the inhibitory effects of FP extract or its fractions in human liver microsomes (HLM), while LC-UV fingerprint and UGT1A1 inhibition profile were combined to identity and characterize the naturally occurring inhibitors of UGT1A1 in FP. n-(3-carboxy propyl)-4-hydroxy-1,8-naphthalimide 13-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 26692528-0 2015 Clinical Implication of UGT1A1 Promoter Polymorphism for Irinotecan Dose Escalation in Metastatic Colorectal Cancer Patients Treated with Bevacizumab Combined with FOLFIRI in the First-line Setting. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26692528-0 2015 Clinical Implication of UGT1A1 Promoter Polymorphism for Irinotecan Dose Escalation in Metastatic Colorectal Cancer Patients Treated with Bevacizumab Combined with FOLFIRI in the First-line Setting. FOLFIRI regimen 164-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26692528-1 2015 PURPOSE: This study aimed to identify the efficacy and toxicity of the FOLFIRI regimen (fluorouracil, leucovorin, and irinotecan) with irinotecan dose escalation plus bevacizumab as first-line chemotherapy for metastatic colorectal cancer (mCRC) via UGT1A1 genotyping. FOLFIRI regimen 71-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 250-256 26692528-1 2015 PURPOSE: This study aimed to identify the efficacy and toxicity of the FOLFIRI regimen (fluorouracil, leucovorin, and irinotecan) with irinotecan dose escalation plus bevacizumab as first-line chemotherapy for metastatic colorectal cancer (mCRC) via UGT1A1 genotyping. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 250-256 26692528-6 2015 RESULTS: The clinical response and disease control rates of mCRC patients treated with FOLFIRI plus bevacizumab were significantly better in patients with UGT1A1 *1/*1 and *1/*28 genotypes than in patients with UGT1A1 *28/*28 (P = .006 and P < .001, respectively). FOLFIRI regimen 87-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 26692528-6 2015 RESULTS: The clinical response and disease control rates of mCRC patients treated with FOLFIRI plus bevacizumab were significantly better in patients with UGT1A1 *1/*1 and *1/*28 genotypes than in patients with UGT1A1 *28/*28 (P = .006 and P < .001, respectively). FOLFIRI regimen 87-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 26342858-3 2015 Bilirubin undergoes selective metabolism by UDP-glucuronosyltransferase (UGT) 1A1 and becomes a water soluble glucuronide. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-81 26429709-7 2015 The study was amended to exclude 4 UGT1A1*28 7/7 homozygous patients due to frequent irinotecan-related grade 3/grade 4 diarrhea and/or febrile neutropenia. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 25649470-0 2015 Interferon and ribavirin combination therapy are linked to severe indirect hyperbilirubinemia in patients with nt-211G > A variant of UGT1A1 gene. Ribavirin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 26543529-4 2015 RESULTS: The mean serum total bilirubin level was significantly higher in the homozygous UGT1A1(AT)7 group as compared to UGT1A1(AT)6 group. Bilirubin 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 26543529-5 2015 Thus, not cholelithiasis but total serum bilirubin was influenced by UGT1A1 polymorphism in this SCA cohort. Bilirubin 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26543529-6 2015 CONCLUSION: As observed in other population groups, the UGT1A1 (AT)7 homozygosity was significantly associated with raised serum total bilirubin level, but the prevalence of gallstones in the Omani SCA patients was not associated with alpha thalassaemia, UGT1A1 polymorphism, or beta(s) haplotypes. Bilirubin 135-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 26482555-8 2015 UGT1A1 genotyping revealed that patients with UGT1A1 polymorphisms exhibited significantly higher response rates to NAC than those with the wild-type (79.5 vs. 49.5 %, respectively, p < 0.05). nac 116-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26482555-8 2015 UGT1A1 genotyping revealed that patients with UGT1A1 polymorphisms exhibited significantly higher response rates to NAC than those with the wild-type (79.5 vs. 49.5 %, respectively, p < 0.05). nac 116-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 26482555-9 2015 CONCLUSIONS: These results indicate that GMP scores of cancerous tissue combined with UGT1A1 genotyping of blood samples may serve as highly potent markers for predicting the efficacy of NAC for individual SCC patients. nac 187-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 26320626-0 2015 Glucuronidation of bavachinin by human tissues and expressed UGT enzymes: Identification of UGT1A1 and UGT1A8 as the major contributing enzymes. bavachinin 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 25988789-3 2015 A series of N-substituted derivatives with various hydrophobic, acidic and basic groups were designed and synthesized to evaluate the selectivity of HN derivatives toward UGT1A1. Nitrogen 12-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 25988789-5 2015 Among the synthesized fluorescent probes, NCHN (N-3-carboxy propyl-4-hydroxy-1,8-naphthalimide) displayed the best combination of selectivity, sensitivity and ratiometric fluorescence response following UGT1A1-catalyzed glucuronidation. nchn 42-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-209 25988789-5 2015 Among the synthesized fluorescent probes, NCHN (N-3-carboxy propyl-4-hydroxy-1,8-naphthalimide) displayed the best combination of selectivity, sensitivity and ratiometric fluorescence response following UGT1A1-catalyzed glucuronidation. n-3-carboxy propyl-4-hydroxy-1,8-naphthalimide 48-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-209 25988789-6 2015 UGT1A1-catalyzed NCHN-4-O-glucuronidation generated a single fluorescent product with a high quantum yield (Phi=0.688) and brought remarkable changes in both color and fluorescence in comparison with the parental substrate. nchn 17-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25988789-6 2015 UGT1A1-catalyzed NCHN-4-O-glucuronidation generated a single fluorescent product with a high quantum yield (Phi=0.688) and brought remarkable changes in both color and fluorescence in comparison with the parental substrate. 4-o 22-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26180128-5 2015 DPF similarly inhibited UGT1A1, UGT1A9, and UGT1A10, with IC50 values ranging from 39 to 66 microM. dapagliflozin 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 26180128-8 2015 Consistent with the activity screening data, DPF was a less potent inhibitor of UGT1A1 and UGT1A9. dapagliflozin 45-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 26180128-9 2015 The Ki for DPF inhibition of UGT1A1 was 81 microM, whereas the Ki values for inhibition of UGT1A9 ranged from 12 to 15 microM. dapagliflozin 11-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 26320626-4 2015 Reaction phenotyping assay showed that UGT1A1, UGT1A3 and UGT1A8 were involved in BCI-4"-O-glucuronidation, while UGT1A1 and UGT1A8 displayed the higher catalytic ability among all tested UGT isoforms. bci-4"-o 82-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 26320626-4 2015 Reaction phenotyping assay showed that UGT1A1, UGT1A3 and UGT1A8 were involved in BCI-4"-O-glucuronidation, while UGT1A1 and UGT1A8 displayed the higher catalytic ability among all tested UGT isoforms. bci-4"-o 82-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 26320626-5 2015 Kinetic analysis demonstrated that BCI-4"-O-glucuronidation in both HLM and UGT1A1 followed sigmoidal kinetic behaviors and displayed much close Km values (12.4 muM in HLM & 9.7 muM in UGT1A1). bci-4"-o 35-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 26320626-5 2015 Kinetic analysis demonstrated that BCI-4"-O-glucuronidation in both HLM and UGT1A1 followed sigmoidal kinetic behaviors and displayed much close Km values (12.4 muM in HLM & 9.7 muM in UGT1A1). bci-4"-o 35-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 26320626-5 2015 Kinetic analysis demonstrated that BCI-4"-O-glucuronidation in both HLM and UGT1A1 followed sigmoidal kinetic behaviors and displayed much close Km values (12.4 muM in HLM & 9.7 muM in UGT1A1). Adenosine Monophosphate 173-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 26320626-6 2015 Both chemical inhibition assays and correlation analysis demonstrated that UGT1A1 displayed a predominant role in BCI-4"-O-glucuronidation in HLM. bci-4"-o 114-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 26320626-9 2015 These findings suggested that UGT1A1 and UGT1A8 were the primary isoforms involved in BCI-4"-O-glucuronidation in HLM, and HIM, respectively. bci-4"-o 86-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 26233886-0 2015 Correlation between UGT1A1 polymorphisms and raltegravir plasma trough concentrations in Japanese HIV-1-infected patients. Raltegravir Potassium 45-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 26233886-1 2015 Raltegravir (RAL), an HIV integrase inhibitor, is metabolized mainly by UDP-glucuronosyltransferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-103 26233886-1 2015 Raltegravir (RAL), an HIV integrase inhibitor, is metabolized mainly by UDP-glucuronosyltransferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 26210671-9 2015 Therefore, nicotine-treated hUGT1 mice might be useful to investigate the role of brain UGT1A3 in the regulation of local levels of these drugs and their response. Nicotine 11-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-33 26163112-8 2015 Glucuronidation of homoegonol to M5 was mediated by UGT1A1, UGT1A3, UGT1A4, and UGT2B7 enzymes, whereas M4 was formed from 4-O-demethylhomoegonol by UGT1A1, UGT1A8, UGT1A10, and UGT2B15 enzymes. homoegonol 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 26163112-8 2015 Glucuronidation of homoegonol to M5 was mediated by UGT1A1, UGT1A3, UGT1A4, and UGT2B7 enzymes, whereas M4 was formed from 4-O-demethylhomoegonol by UGT1A1, UGT1A8, UGT1A10, and UGT2B15 enzymes. homoegonol 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 26413716-15 2015 In addition, our findings indicate that similar to the adult population, the UGT1A1 is the main locus responsible for normal variation of serum bilirubin in pediatric populations. Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 25782327-0 2015 DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. fluoropyrimidines 92-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 25782327-0 2015 DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Oxaliplatin 111-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 25782327-0 2015 DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 25782327-2 2015 Single nucleotide polymorphisms (SNPs) in DPYD and UGT1A1 influence fluoropyrimdines and irinotecan adverse events (AEs). fluoropyrimdines 68-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 25782327-2 2015 Single nucleotide polymorphisms (SNPs) in DPYD and UGT1A1 influence fluoropyrimdines and irinotecan adverse events (AEs). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 26072390-8 2015 Poor responders had a higher conjugated/unconjugated bilirubin ratio, indicating increased UGT1A1 activity. Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 26247603-10 2015 There are human-specific changes in the sequence of the promoter and the protein-coding sequence of uridine diphosphoglucuronosyltransferase 1 (UGT1A1), the enzyme that transforms bilirubin and toxic plant compounds into water-soluble, excretable metabolites. Bilirubin 180-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 26247603-10 2015 There are human-specific changes in the sequence of the promoter and the protein-coding sequence of uridine diphosphoglucuronosyltransferase 1 (UGT1A1), the enzyme that transforms bilirubin and toxic plant compounds into water-soluble, excretable metabolites. Water 221-226 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 26247603-11 2015 Experimental evidence that UGT1A1 is down-regulated in the human liver suggests that changes in the promoter may be responsible for the human-specific increase in bilirubin. Bilirubin 163-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 26247603-12 2015 We speculate that since cooking reduces toxic plant compounds, consumption of cooked foods, which is specific to humans, may have resulted in relaxed constraint on UGT1A1 which has in turn led to higher serum levels of bilirubin in humans. Bilirubin 219-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 25611851-9 2015 Furthermore, we reported our present work on genetic analysis of mutations of UGT1A1 gene, and the correlation of UGT1A1 mutations with serum total bilirubin levels in Taiwanese population. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 26229432-0 2015 Relationship between UGT1A1*6/*28 polymorphisms and severe toxicities in Chinese patients with pancreatic or biliary tract cancer treated with irinotecan-containing regimens. Irinotecan 143-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 26229432-1 2015 PURPOSE: The aim of this retrospective study was to investigate the relationship between UGT1A1 polymorphisms and toxicities in Chinese patients with pancreatic or biliary tract cancer receiving irinotecan-containing regimens as the second- or third-line chemotherapy. Irinotecan 195-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 26229432-12 2015 CONCLUSION: In Chinese patients with pancreatic or biliary tract cancer administered irinotecan-containing regimens, those with UGT1A1*6 variant may have a high risk of severe neutropenia. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 26146841-3 2015 Eleven common mutations and polymorphisms across five bilirubin metabolism genes, namely those encoding UGT1A1, HMOX1, BLVRA, SLCO1B1 and SLCO1B3, were determined using the high resolution melt (HRM) assay or PCR-capillary electrophoresis analysis. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 26066637-0 2015 Decreased Expression of Multidrug Resistance-Associated Protein 4 (MRP4/ABCC4) Leads to Reduced Glucuronidation of Flavonoids in UGT1A1-Overexpressing HeLa Cells: The Role of Futile Recycling. Flavonoids 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 25870101-0 2015 Glucuronidation of OTS167 in Humans Is Catalyzed by UDP-Glucuronosyltransferases UGT1A1, UGT1A3, UGT1A8, and UGT1A10. OTS167 19-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 25870101-6 2015 Nilotinib (UGT1A1 inhibitor) and emodin (UGT1A8 and UGT1A10 inhibitor) exhibited the highest inhibitory effects on OTS167-G formation in HLM (68%) and HIM (47%). nilotinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 26180834-0 2015 Screening for UGT1A1 Genotype in Study A5257 Would Have Markedly Reduced Premature Discontinuation of Atazanavir for Hyperbilirubinemia. Atazanavir Sulfate 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 26180834-3 2015 Atazanavir-associated hyperbilirubinemia correlates with UGT1A1 rs887829 genotype. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 26180834-6 2015 Discriminatory properties of UGT1A1 T/T genotype for predicting bilirubin-related atazanavir discontinuation through 96 weeks after antiretroviral initiation were estimated. Bilirubin 64-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 26180834-6 2015 Discriminatory properties of UGT1A1 T/T genotype for predicting bilirubin-related atazanavir discontinuation through 96 weeks after antiretroviral initiation were estimated. Atazanavir Sulfate 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 26591441-0 2015 [UGT1A1 Genotyping for Proper Use of Irinotecan]. Irinotecan 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 1-7 26591441-3 2015 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by the UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-135 26591441-3 2015 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by the UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-135 26591441-4 2015 Recent pharmacogenetic studies on irinotecan have revealed the impact of UGT1A1 polymorphisms on severe adverse effects. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 26591441-5 2015 A variant in the promoter of the UGT1A1 gene, the UGT1A1 *28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and severe toxicities of irinotecan have been reported. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-5 2015 A variant in the promoter of the UGT1A1 gene, the UGT1A1 *28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and severe toxicities of irinotecan have been reported. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 26591441-8 2015 The concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, markedly alters the disposition of irinotecan, potentially increasing toxicity, which is now written on the label of irinotecan in Japan. Irinotecan 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26591441-9 2015 For patients showing homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 26591441-9 2015 For patients showing homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28, dose reduction of irinotecan is strongly recommended. Irinotecan 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 26591441-11 2015 Moreover, a recent Phase 2 trial for FOLFIRINOX in Japan excluded patients who showed homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28. folfirinox 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 26591441-11 2015 Moreover, a recent Phase 2 trial for FOLFIRINOX in Japan excluded patients who showed homozygosity for UGT1A1*28, *6, or compound heterozygosity for UGT1A1*6 and *28. folfirinox 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 26591441-12 2015 At present, irinotecan chemotherapy based on a patient"s UGT1A1 genetic status is scientifically reasonable. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 25834030-7 2015 We found that zafirlukast is a potent general inhibitor of UGT1As and a moderate inhibitor of UGT2Bs because it monitors 4-methylumbelliferone glucuronidation by recombinant UGTs. zafirlukast 14-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-63 26125934-0 2015 Correlation of UGT1A1 and ERCC1 gene polymorphisms with the outcome of combined irinotecan plus cisplatin treatment in recurrent ovarian cancer. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26125934-0 2015 Correlation of UGT1A1 and ERCC1 gene polymorphisms with the outcome of combined irinotecan plus cisplatin treatment in recurrent ovarian cancer. Cisplatin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 26125934-1 2015 The aim of this study was to define the genotypes of UGT1A1 and ERCC1 and to examine their relationship with the efficacy and toxicity of a combination therapy of irinotecan and cisplatin in patients with advanced ovarian cancer. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 26125934-1 2015 The aim of this study was to define the genotypes of UGT1A1 and ERCC1 and to examine their relationship with the efficacy and toxicity of a combination therapy of irinotecan and cisplatin in patients with advanced ovarian cancer. Cisplatin 178-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 26125934-7 2015 Together, the results from this study suggest that UGT1A1 is a target gene for tardive diarrhea, and that the UGT1A1*28 gene mutation might increase the risk of diarrhea with irinotecan-based chemotherapy. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 26125934-7 2015 Together, the results from this study suggest that UGT1A1 is a target gene for tardive diarrhea, and that the UGT1A1*28 gene mutation might increase the risk of diarrhea with irinotecan-based chemotherapy. Irinotecan 175-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 25917289-4 2015 Our data indicated that both gefitinib and DMG are potent inhibitors of SN-38 glucuronidation via UGT1A1 inhibition. Gefitinib 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 25917289-4 2015 Our data indicated that both gefitinib and DMG are potent inhibitors of SN-38 glucuronidation via UGT1A1 inhibition. dimethylglycine 43-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 25917289-4 2015 Our data indicated that both gefitinib and DMG are potent inhibitors of SN-38 glucuronidation via UGT1A1 inhibition. Irinotecan 72-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 25315738-6 2015 Uridine diphospho-glucuronosyl transferase 1A1 (UGT1A1) is responsible for the glucuronidation of bilirubin; deficiency of this enzyme results in unconjugated hyperbilirubinaemia. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-46 25315738-6 2015 Uridine diphospho-glucuronosyl transferase 1A1 (UGT1A1) is responsible for the glucuronidation of bilirubin; deficiency of this enzyme results in unconjugated hyperbilirubinaemia. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 25817555-4 2015 The present review on the impact of the deficient UGT1A1*28 variant on irinotecan efficacy and toxicity was produced by a French joint workgroup comprising the Group of Clinical Onco-pharmacology (GPCO-Unicancer) and the National Pharmacogenetics Network (RNPGx). Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25817555-5 2015 It clearly emerges that for irinotecan doses at least equal to 180 mg/m(2) , patients homozygous for the UGT1A1*28 allele are at increased risk of developing hematological and/or digestive toxicities. Irinotecan 28-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25081908-11 2015 A significant inverse correlation (P = 0.03) was observed between CDFX and UGT1A1*28 gene polymorphism, but only in patients with levels of lean body mass (LBM) below the median (P for interaction = 0.05). cdfx 66-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 26039129-11 2015 UGT1A1 intronic single-nucleotide polymorphisms (SNPs) (rs6742078, rs887829, rs4148324) serve as proxy markers for the low-frequency and rare UGT1A1 variants, thereby providing mechanistic explanation to the relationship between UGT1A1 intronic SNPs and the UGT1A1 enzyme activity. single-nucleotide 16-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26039129-11 2015 UGT1A1 intronic single-nucleotide polymorphisms (SNPs) (rs6742078, rs887829, rs4148324) serve as proxy markers for the low-frequency and rare UGT1A1 variants, thereby providing mechanistic explanation to the relationship between UGT1A1 intronic SNPs and the UGT1A1 enzyme activity. single-nucleotide 16-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 26039129-11 2015 UGT1A1 intronic single-nucleotide polymorphisms (SNPs) (rs6742078, rs887829, rs4148324) serve as proxy markers for the low-frequency and rare UGT1A1 variants, thereby providing mechanistic explanation to the relationship between UGT1A1 intronic SNPs and the UGT1A1 enzyme activity. single-nucleotide 16-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 26039129-11 2015 UGT1A1 intronic single-nucleotide polymorphisms (SNPs) (rs6742078, rs887829, rs4148324) serve as proxy markers for the low-frequency and rare UGT1A1 variants, thereby providing mechanistic explanation to the relationship between UGT1A1 intronic SNPs and the UGT1A1 enzyme activity. single-nucleotide 16-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 25993113-3 2015 In East Asian populations, the compound homozygous UGT1A1 G71R and Y486D variants are frequently observed in cases with bilirubin levels exceeding 200 mumol/L. Bilirubin 120-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 25993113-8 2015 Therefore, we propose that the spectrum of UGT1A1 variations in CNS-II differs according to the bilirubin levels. Bilirubin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 200-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25880011-1 2015 BACKGROUND: In Asians, the risk of irinotecan-induced severe toxicities is related in part to UGT1A1*6 (UGT, UDP glucuronosyltransferase) and UGT1A1*28, variant alleles that reduce the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 232-242 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25880011-2 2015 We prospectively studied the relation between the UGT1A1 genotype and the safety of irinotecan-based regimens in Japanese patients with advanced colorectal cancer, and then constructed a nomogram for predicting the risk of severe neutropenia in the first treatment cycle. Irinotecan 84-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25880011-6 2015 RESULTS: The UGT1A1 genotype was confirmed to be associated with increased risks of irinotecan-induced grade 3 or 4 neutropenia and diarrhoea. Irinotecan 84-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25966095-1 2015 Mutations in the UGT1A1 gene cause Crigler-Najjar syndrome (CN), which causes non-hemolytic unconjugated hyperbilirubinemia, and is categorized as CN1 and CN2 according to the severity of bilirubin levels. Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 25595598-0 2015 Characterization of chrysin glucuronidation in UGT1A1-overexpressing HeLa cells: elucidating the transporters responsible for efflux of glucuronide. Glucuronides 136-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 25595598-5 2015 Ko143 at 10-20 muM (a dual inhibitor of BCRP and UGT1A1) caused a marked decrease (51.3%-59.7%, P < 0.01) in the excretion rate and efflux clearance of CG. 3-(6-isobutyl-9-methoxy-1,4-dioxo-1,2,3,4,6,7,12,12a-octahydropyrazino(1',2'-1,6)pyrido(3,4-b)indol-3-yl)propionic acid tert-butyl ester 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 25595598-5 2015 Ko143 at 10-20 muM (a dual inhibitor of BCRP and UGT1A1) caused a marked decrease (51.3%-59.7%, P < 0.01) in the excretion rate and efflux clearance of CG. cg 155-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 25595598-6 2015 Likewise, MK-571 at 5-20 muM (an inhibitor of MRPs but an activator of UGT1A1) resulted in a significant reduction in the excretion rate (18.2%-64.0%, P < 0.01) and efflux clearance (37.0%-90.2%, P < 0.001). verlukast 10-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 26039129-6 2015 These variants were in strong linkage disequilibrium with 3 intronic UGT1A1 variants (rs887829, rs4148325, rs6742078), which were significantly associated with total bilirubin level (P = 2.34 x 10(-34), P = 7.02 x 10(-34), and P = 8.27 x 10(-34)), as well as unconjugated, and conjugated bilirubin levels. Bilirubin 166-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26039129-6 2015 These variants were in strong linkage disequilibrium with 3 intronic UGT1A1 variants (rs887829, rs4148325, rs6742078), which were significantly associated with total bilirubin level (P = 2.34 x 10(-34), P = 7.02 x 10(-34), and P = 8.27 x 10(-34)), as well as unconjugated, and conjugated bilirubin levels. Bilirubin 288-297 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26039129-10 2015 Using an exome-wide approach we identified coding variants on UGT1A1 and UGT1A6 genes in association with serum bilirubin level and hyperbilirubinemia risk in elderly subjects. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25348619-2 2015 Our aim was to evaluate DFX plasma concentrations according to single-nucleotide polymorphisms in genes involved in its metabolism (UGT1A1, UGT1A3, CYP1A1, CYP1A2 and CYP2D6) and elimination (MRP2 and BCRP1). Deferasirox 24-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 25522350-6 2015 Diflunisal inhibited the UGT1A1 (IC50 = 33.0 mum) and UGT1A9 (IC50 = 19.4 mum). Diflunisal 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 25981652-2 2015 However, there are reports of an association between certain UGT1A1 genetic polymorphisms and the development of adverse reactions(such as neutropenia)related to irinotecan metabolism. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25981652-7 2015 These results suggest that a dose reduction of irinotecan should be considered for patients who fall into the homozygous group upon analysis of their UGT1A1 genetic polymorphisms, as such patients might be susceptible to grade 4 neutropenia. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 25081908-14 2015 Interesting results emerged from the analysis of genetic and physical characteristics of patients: LBM was a borderline significant effect modifier of the relationship between UGT1A1 polymorphisms and CDFX . cdfx 201-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 25663682-7 2015 Haploinsufficiency at MLXIPL and increased penetrance for hypomorphic alleles at the UGT1A1 gene promoter might underlie the lipid and bilirubin alterations. Bilirubin 135-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 25171434-0 2015 UGT1A1*28 is associated with greater decrease in serum K+ levels following oral intake of procaterol. Procaterol 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25171434-8 2015 Meanwhile, overall serum K(+) level changes were significantly lower in carriers of UGT1A1*28 than in non-carriers after correcting for strong effects of serum procaterol concentrations and baseline K(+) levels. Procaterol 160-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 25171434-11 2015 CONCLUSION: The present study indicates that significant hypokalemia may occur in carriers of UGT1A1*28 by systemic administration of procaterol and potentially by other beta2-agonists metabolized in the liver. Procaterol 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 25834402-16 2015 CONCLUSION: In mCRC patients with wild-type or heterozygous UGT1A1*6 or *28 genotype, biweekly XELIRI + BV is effective and feasible as second-line chemotherapy. xeliri + bv 95-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 26375741-5 2015 The relationship between A(TA)nTAA variation of UGT1A1 gene, the serum bilirubin level and the occurrence of cholilithiasis was investigated. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 26375741-5 2015 The relationship between A(TA)nTAA variation of UGT1A1 gene, the serum bilirubin level and the occurrence of cholilithiasis was investigated. cholilithiasis 109-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 24905729-11 2015 In vitro, rifampicin PXR-dependently upregulated biotransformation phase 1 (CYP3A4), phase 2 (UGT1A1) and phase 3 (MRP2) enzymes/carriers as well as the basolateral bile salt exporter OSTbeta. Rifampin 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 24905729-14 2015 PXR-dependent induction of CYP3A4, UGT1A1, MRP2 and OSTbeta could contribute to the anticholestatic effect of rifampicin in PHSF. Rifampin 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 26012253-5 2015 Inhibition kinetic type and parameter (Ki) were determined for the inhi- bition of podophyllotoxin towards UGT1A1, and competitive inhibition of podophyllotoxin towards UGT1A1 was observed with the inhibition kinetic parameter (Ki) to be 4.0 muM. Podophyllotoxin 83-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 26012253-5 2015 Inhibition kinetic type and parameter (Ki) were determined for the inhi- bition of podophyllotoxin towards UGT1A1, and competitive inhibition of podophyllotoxin towards UGT1A1 was observed with the inhibition kinetic parameter (Ki) to be 4.0 muM. Podophyllotoxin 145-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 26012253-7 2015 In conclusion, podophyllotoxin inhibited UGT1A1 activity, indicating potential herb-drug interactions between podophyllotoxin-containing herbs and drugs mainly undergoing UGT1A1-mediated metabolism. Podophyllotoxin 15-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 26012253-7 2015 In conclusion, podophyllotoxin inhibited UGT1A1 activity, indicating potential herb-drug interactions between podophyllotoxin-containing herbs and drugs mainly undergoing UGT1A1-mediated metabolism. Podophyllotoxin 15-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 26012253-7 2015 In conclusion, podophyllotoxin inhibited UGT1A1 activity, indicating potential herb-drug interactions between podophyllotoxin-containing herbs and drugs mainly undergoing UGT1A1-mediated metabolism. Podophyllotoxin 110-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 26012253-7 2015 In conclusion, podophyllotoxin inhibited UGT1A1 activity, indicating potential herb-drug interactions between podophyllotoxin-containing herbs and drugs mainly undergoing UGT1A1-mediated metabolism. Podophyllotoxin 110-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 25596428-2 2015 DES can potently inhibit E2-3-O-glucuronidation in HLM, a probe reaction for UGT1A1. Diethylstilbestrol 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 25611302-2 2015 The active metabolite of irinotecan, SN-38, is a known substrate of drug-metabolising enzymes, including UGT1A1, as well as OATP and ABC drug transporters. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25576848-2 2015 Gilbert"s Syndrome is associated with mutation in the Uridine Glucuronosyl Transferase 1A1 (UGT1A1) gene promoter, reducing UGT1A1 activity, which normally conjugates bilirubin allowing its elimination from the blood. Bilirubin 167-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-90 25576848-2 2015 Gilbert"s Syndrome is associated with mutation in the Uridine Glucuronosyl Transferase 1A1 (UGT1A1) gene promoter, reducing UGT1A1 activity, which normally conjugates bilirubin allowing its elimination from the blood. Bilirubin 167-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 25576848-2 2015 Gilbert"s Syndrome is associated with mutation in the Uridine Glucuronosyl Transferase 1A1 (UGT1A1) gene promoter, reducing UGT1A1 activity, which normally conjugates bilirubin allowing its elimination from the blood. Bilirubin 167-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 25858605-4 2015 Dolutegravir is metabolized by UGT1A1 and, to a lesser extent, by CYP3A4, without being an inducer or inhibitor of the usual metabolic systems. dolutegravir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 25611302-2 2015 The active metabolite of irinotecan, SN-38, is a known substrate of drug-metabolising enzymes, including UGT1A1, as well as OATP and ABC drug transporters. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 25611302-5 2015 RESULTS: SLCO1B1 521C was significantly associated with increased SN-38 exposure (P<0.001), which was additive with UGT1A1*28. Irinotecan 66-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 25596428-10 2015 In summary, this study issues new potential toxic mechanisms for DES: potently inhibiting the activity of UGT1A1 and powerfully accelerating the formation of cholestatic E2-17-O-glucuronide by UGT1A4. Diethylstilbestrol 65-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 25052959-4 2015 HS-23 showed negligible inhibition of CYP1A2, CYP2C8, CYP2D6, UGT1A1, UGT1A4, UGT1A9, and UGT2B7 activities in human liver microsomes. hs-23 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25692465-0 2015 UDP-glucuronosyltransferase 1A determinates intracellular accumulation and anti-cancer effect of beta-lapachone in human colon cancer cells. beta-lapachone 97-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. 7-ethyl-10-hydroxycamptothecin glucuronide 150-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. 7-ethyl-10-hydroxycamptothecin glucuronide 150-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25689738-4 2015 In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 25760534-0 2015 UDP-glucuronosyltransferase (UGT) 1A1 mainly contributes to the glucuronidation of trovafloxacin. trovafloxacin 83-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 25760534-5 2015 In human liver microsomes, trovafloxacin acyl-glucuronidation followed the Hill equation with S50 value of 95 muM, Vmax value of 243 pmol/min per mg, and a Hill coefficient of 2.0, while the UGT1A1-expressing system displayed Michaelis-Menten kinetics with a substrate inhibition, with Km value of 759 muM and Vmax value of 1160 pmol/min per mg. trovafloxacin 27-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. trovafloxacin 97-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. trovafloxacin 97-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 25760534-3 2015 In the present study, among the functional human UGT members, UGT1A1, UGT1A3, and UGT1A9 exhibited higher trovafloxacin acyl-glucuronidation activities. trovafloxacin 106-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. acyl-glucuronide 111-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. acyl-glucuronide 111-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 25997248-0 2015 UGT1A1*28 polymorphism influences glucuronidation of bazedoxifene. bazedoxifene 53-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. trovafloxacin 291-304 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 25760534-6 2015 In human liver microsomes prepared from poor metabolizers (UGT1A1*28/*28), significantly reduced trovafloxacin acyl-glucuronide formation activity was observed, indicating that UGT1A1 mainly, while other UGT members such as UGT1A3 and UGT1A9 partially, contributes to the glucuronidation of trovafloxacin. trovafloxacin 291-304 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 25997248-3 2015 It has already been shown that a relatively common UGT1A1*28 polymorphism can considerably affect raloxifene pharmacokinetics and pharmacodynamics. Raloxifene Hydrochloride 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 25997248-4 2015 As pharmacokinetics of bazedoxifene and raloxifene are very similar, the influence of UGT1A1*28 polymorphism on metabolism of bazedoxifene was investigated by genotyped microsomes. bazedoxifene 126-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 25997248-5 2015 Our results indicate an influence of UGT1A1*28 allele on the formation clearance of both bazedoxifene metabolites. bazedoxifene 89-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 25997248-7 2015 In conclusion, the significant UGT1A1*28 genotype effect on bazedoxifene intrinsic metabolic clearance indicates that this subject is worth further exploration in vivo and provides valuable information research in this field. bazedoxifene 60-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 25055799-0 2015 UGT1A1 *6 polymorphism predicts outcome in elderly patients with relapsed or refractory diffuse large B-cell lymphoma treated with carboplatin, dexamethasone, etoposide and irinotecan. Carboplatin 131-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25598834-2 2015 RAL should be double-dosed to 800 mg semi-daily in situation of rifampicin co-medication, because RAL is more rapidly metabolized due to rifampicin-induced Uridine-5"-diphosph- gluronosyl-transferase (UGT1A1). Rifampin 137-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 25055799-3 2015 The 2-year survival rate after initial CDE-11 treatment was significantly higher in patients with than without UGT1A1*6 (57% vs. 5%). cde-11 39-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 25055799-0 2015 UGT1A1 *6 polymorphism predicts outcome in elderly patients with relapsed or refractory diffuse large B-cell lymphoma treated with carboplatin, dexamethasone, etoposide and irinotecan. Dexamethasone 144-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25055799-7 2015 The response to CDE-11 likely correlated with UGT1A1*6 polymorphisms, but further prospective studies are warranted to optimize irinotecan-based chemotherapies relative to UGT1A1 polymorphism. cde-11 16-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 25055799-0 2015 UGT1A1 *6 polymorphism predicts outcome in elderly patients with relapsed or refractory diffuse large B-cell lymphoma treated with carboplatin, dexamethasone, etoposide and irinotecan. Etoposide 159-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25055799-0 2015 UGT1A1 *6 polymorphism predicts outcome in elderly patients with relapsed or refractory diffuse large B-cell lymphoma treated with carboplatin, dexamethasone, etoposide and irinotecan. Irinotecan 173-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25845873-3 2015 From the interaction between atazanavir and DTG via CYP3A4 and UGT1A1 and placental efflux transporter inhibition and considering the infant"s probable enzymatic immaturity, the DTG elimination half-life was estimated to be 4-fold longer in neonates than in adults. Atazanavir Sulfate 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 25385099-2 2015 Faldaprevir is known to inhibit P-glycoprotein, CYP3A4, and UDP-glucuronosyltransferase 1A1. faldaprevir 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-91 25845873-3 2015 From the interaction between atazanavir and DTG via CYP3A4 and UGT1A1 and placental efflux transporter inhibition and considering the infant"s probable enzymatic immaturity, the DTG elimination half-life was estimated to be 4-fold longer in neonates than in adults. dolutegravir 44-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 25808438-5 2015 For upfront identification of patients at high risk of suffering from severe therapy-related toxicity, specific variants of dihydropyrimidine dehydrogenase may be measured for predicting toxicity from fluoropyrimidines and uridine diphosphate glucuronosyltransferase*28 (UGT1A1*28) for predicting toxicity from irinotecan. fluoropyrimidines 201-218 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 26632182-2 2015 UGT1A1 enzyme activity is commonly evaluated by detecting the elimination of bilirubin substrate or the generation of bilirubin glucuronides. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26632182-2 2015 UGT1A1 enzyme activity is commonly evaluated by detecting the elimination of bilirubin substrate or the generation of bilirubin glucuronides. Bilirubin 118-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 26632182-2 2015 UGT1A1 enzyme activity is commonly evaluated by detecting the elimination of bilirubin substrate or the generation of bilirubin glucuronides. Glucuronides 128-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25947908-6 2015 For patients with a UGT1A1*6/*6, *6/*28, or *28/*28 genotype initially administered 300-400 mg/d, a target nilotinib C0 of 500 ng/mL is recommended to prevent elevation of bilirubin levels, whereas for patients with the UGT1A1*1 allele initially administered 600 mg/d, a target nilotinib C0 of 800 ng/mL is recommended. nilotinib c0 107-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 26087609-0 2015 [The role of assessing UGT1A1 gene polymorphism in the prediction of irinotecan-induced toxicity in the course of chemotherapy for colorectal cancer]. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 25427841-0 2015 A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 25427841-0 2015 A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI. Irinotecan 73-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 25427841-0 2015 A phase I study of UGT1A1 *28/*6 genotype-directed dosing of irinotecan (CPT-11) in Korean patients with metastatic colorectal cancer receiving FOLFIRI. FOLFIRI regimen 144-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 25427841-1 2015 PURPOSE: A UGT1A1 genotype-directed dose escalation of irinotecan (CPT-11) was performed in patients with metastatic colorectal cancer receiving first-line FOLFIRI chemotherapy. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 25427841-1 2015 PURPOSE: A UGT1A1 genotype-directed dose escalation of irinotecan (CPT-11) was performed in patients with metastatic colorectal cancer receiving first-line FOLFIRI chemotherapy. Irinotecan 67-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 25427841-8 2015 CONCLUSIONS: The MTD of irinotecan differed according to the UGT1A1 genotype, and higher doses of irinotecan are feasible with sLV5FU2 compared to the present regulatory approved doses. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25427841-8 2015 CONCLUSIONS: The MTD of irinotecan differed according to the UGT1A1 genotype, and higher doses of irinotecan are feasible with sLV5FU2 compared to the present regulatory approved doses. Irinotecan 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 25714435-7 2015 Enzyme kinetic studies showed that UGT1A1 was the main UGT isoform involved in OCG in HLMs. CHEMBL1234931 79-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 25475995-10 2015 Characterization of UGT isoenzymes in glucuronidation of tanzisertib and CC-418424 revealed that, tanzisertib glucuronide was catalyzed by: UGT1A1, 1A4, 1A10 and 2B4, while CC-418424 glucuronidation was catalyzed by UGT2B4 and 2B7. 4-(9-(tetrahydrofuran-3-yl)-8-(2,4,6-trifluorophenylamino)-9H-purin-2-ylamino)cyclohexanol 57-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 25475995-10 2015 Characterization of UGT isoenzymes in glucuronidation of tanzisertib and CC-418424 revealed that, tanzisertib glucuronide was catalyzed by: UGT1A1, 1A4, 1A10 and 2B4, while CC-418424 glucuronidation was catalyzed by UGT2B4 and 2B7. cc-418424 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 25475995-10 2015 Characterization of UGT isoenzymes in glucuronidation of tanzisertib and CC-418424 revealed that, tanzisertib glucuronide was catalyzed by: UGT1A1, 1A4, 1A10 and 2B4, while CC-418424 glucuronidation was catalyzed by UGT2B4 and 2B7. tanzisertib glucuronide 98-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 25751380-5 2015 RESULTS: Sequencing of the Gilbert syndrome-associated gene, UGT 1A 1, revealed mutations in the upstream promoter phenobarbital-responsive element module (PBREM) (-3279 mutation, 23 cases), in the promoter TATA box (a TA insertion mutation, 21 cases), and in the coding region of exon 1 (a GGA-AGA Gly71Arg mutation, 18 cases); there was no statistical difference found for any of the three mutations among this patient population (x2 =1.640, P more than 0.05). Phenobarbital 115-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-69 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 26068529-2 2015 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an irreplaceable role in detoxification of bilirubin and many drugs (e.g., SN-38). Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Atazanavir Sulfate 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 26068529-5 2015 Determination of glucuronidation rates (beta-estradiol and SN-38 as the substrates) was performed using HeLa1A1 cells and uridine diphosphoglucuronic acid (UDPGA)-supplemented cDNA expressed UGT1A1 enzyme (or microsomes). Uridine Diphosphate Glucuronic Acid 122-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 25202035-1 2014 OBJECTIVE: To report a case of systemic irinotecan toxicity following regional transarterial chemoembolization with drug-eluting beads loaded with irinotecan (DEBIRI-TACE) in a patient later found to have a homozygous mutation for UGT1A1*28. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 25202035-7 2014 Patients with genetic polymorphisms of the genes encoding for the enzyme UGT1A1 may have increased incidence of irinotecan-associated toxicities because of decreased clearance of the active metabolite SN38 via the glucuronidation pathway. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 25217485-5 2014 Evaluation of PRAG formation by recombinant UDP-glucuronosyltransferase (UGT) isoforms and an inhibition study using HLHs as an enzyme source demonstrated that multiple UGT isoforms, including UGT1A1, UGT1A9, and UGT2B7, catalyzed probenecid acyl glucuronidation. Probenecid 231-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 25877315-0 2015 [Association of UGT1A1 (*28, *60 and * 93) polymorphism with the adverse reactions of irinotecan chemotherapy in extensive stage small cell lung cancer]. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 25877315-1 2015 OBJECTIVE: To explore the correlation between UGT1A1 (*28, *60 and * 93) polymorphism and the adverse reactions in small cell lung cancer patients after irinotecan chemotherapy. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 25877315-9 2015 Further prospective study in a larger number of patients is needed to clarify the association between UGT1A1*28, UGT1A1* 93 and UGT1A1*60 polymorphism and adverse reactions of irinotecan, and to help clinicians in choosing a better therapeutic modality for personalized chemotherapy to improve curative effect and reduce adverse reactions. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 25151207-3 2014 METHODS: Nineteen SNPs in the ABCB1, NR1I2, UGT1A1, SLCO1B1 and CYP3A5 genes were examined in case patients with atazanavir/ritonavir-induced nephrolithiasis (n = 31) and controls (n = 47). Ritonavir 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 25312789-6 2014 Of these subjects, 21% required a warfarin dose higher than 70 mg/week, which was associated with a BMI greater than 25 kg/m(2), use of warfarin antagonists, and the presence of the MDR1 3435T allele and UGT1A1(TA) 7 polymorphism. Warfarin 34-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 204-210 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Bilirubin 262-271 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 25262300-7 2014 CONCLUSION: Bilirubin levels and changes during the middle and late parts of the first week were attributed to variants and haplotypes in UGT1A1. Bilirubin 12-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-70 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-244 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). thymine-adenine 167-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-70 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). thymine-adenine 167-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 25086287-2 2014 The enzyme uridine diphosphate glucuronosyltransferase polypeptide 1A1 (UGT1A1) is solely responsible for clearing bilirubin from the blood and homozygosity for seven thymine-adenine (TA) repeats in the TATA box regulatory element of the UGT1A1 gene underlies a mild hereditary unconjugated hyperbilirubinaemia (Gilbert"s syndrome). thymine-adenine 167-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-244 25262300-1 2014 OBJECTIVE: To explore the effects of variants in Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) and Heme Oxygenase-1 (HMOX1) on daily physiological bilirubin levels and bilirubin changes during the first week after birth in Chinese newborns. Bilirubin 159-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-97 25262300-1 2014 OBJECTIVE: To explore the effects of variants in Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) and Heme Oxygenase-1 (HMOX1) on daily physiological bilirubin levels and bilirubin changes during the first week after birth in Chinese newborns. Bilirubin 159-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 25262300-2 2014 Both UGT1A1 and HMOX1 code rate-limiting enzymes in the bilirubin metabolism pathway. Bilirubin 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Bilirubin 104-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Bilirubin 104-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Carbon 76-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Carbon 76-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 25262300-4 2014 RESULTS: For UGT1A1, we found minor allele A of rs4148323 (G211A, UGT1A1*6) contributed to higher daily bilirubin levels on days 4-6 (with contributions to variations increasing from 4.8% to 12.3%), minor allele T of rs887829 (c-364t) contributed to lower daily bilirubin levels for days 6 and 7 (with contributions to variations increasing from 7.0% to 10.2%) (P < .03 for all). Bilirubin 262-271 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25312789-9 2014 The UGT1A1(TA) 7 allele was positively correlated with the warfarin dose. Warfarin 59-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 25312789-10 2014 CONCLUSION: CYP2C9*2, CYP2C9*3, VKORC1-1639G>A, and APOE *epsilon4 were associated with lower warfarin doses, while MDR1 3435C>T and UGT1A1(TA) n polymorphisms were associated with a requirement for higher doses. Warfarin 97-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 25364403-3 2014 The current study demonstrates that rapamycin may enhance the chemopreventive effects of SFN on Caco-2 cells; this may be partially attributed to nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2)- and human pregnane X receptor (hPXR)-mediated UGT1A1, UGT1A8 and UGT1A10 induction. Sirolimus 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 269-275 25394051-4 2014 Atazanavir (ATV) inhibits UGT1A1, thus increasing unconjugated BR levels, a distinctive feature of this drug. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 25473295-0 2014 FOLFIRI and regorafenib combination therapy with dose escalation of irinotecan as fourth-line treatment for patients with metastatic colon cancer according to UGT1A1 genotyping. FOLFIRI regimen 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 25200497-1 2014 BACKGROUNDS AND AIMS: UDP-glucuronosyltransferase 1 A1 (UGT1A1) is an enzyme that transforms small lipophilic molecules into water-soluble and excretable metabolites. Water 125-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-54 25200497-1 2014 BACKGROUNDS AND AIMS: UDP-glucuronosyltransferase 1 A1 (UGT1A1) is an enzyme that transforms small lipophilic molecules into water-soluble and excretable metabolites. Water 125-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 181-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 181-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 181-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-9 2014 Quantitative analysis showed that sense mutation (including UGT1A1*6) and UGT1A1*28/*28, but not UGT1A1*60/*60 or UGT1A1*1/*28, was associated with increased serum total bilirubin (TB) levels. Bilirubin 181-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 25200497-11 2014 CONCLUSIONS: This study identified four novel UGT1A1 coding variants, some of which were associated with increased serum TB levels. Bilirubin 121-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 25394086-4 2014 ATV contributes to the decreased levels of UGT1A1, which may lead to elevations of indirect bilirubin, jaundice and even to therapy discontinuation. Atazanavir Sulfate 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 25394086-4 2014 ATV contributes to the decreased levels of UGT1A1, which may lead to elevations of indirect bilirubin, jaundice and even to therapy discontinuation. Bilirubin 92-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 25394086-19 2014 CONCLUSIONS: The risk of severe hyperbilirubinemia during ATV treatment is minimal for patients without UGT1A1*28 and no more than one additional risk factor and for patients with UGT1A1*28 and no additional risk factors; patients with homozygous genotype UGT1A1*28 are at the highest risk. Atazanavir Sulfate 58-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 25394086-19 2014 CONCLUSIONS: The risk of severe hyperbilirubinemia during ATV treatment is minimal for patients without UGT1A1*28 and no more than one additional risk factor and for patients with UGT1A1*28 and no additional risk factors; patients with homozygous genotype UGT1A1*28 are at the highest risk. Atazanavir Sulfate 58-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 25394086-19 2014 CONCLUSIONS: The risk of severe hyperbilirubinemia during ATV treatment is minimal for patients without UGT1A1*28 and no more than one additional risk factor and for patients with UGT1A1*28 and no additional risk factors; patients with homozygous genotype UGT1A1*28 are at the highest risk. Atazanavir Sulfate 58-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 25192553-0 2014 Exploring the roles of UGT1A1 and UGT1A3 in oral clearance of GSK2190915, a 5-lipoxygenase-activating protein inhibitor. 3-(3-tert-butylsulfanyl-1-(4-(6-ethoxypyridin-3-yl)benzyl)-5-(5-methylpyridin-2-ylmethoxy)-1H-indol-2-yl)-2,2-dimethylpropionic acid 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 25473295-0 2014 FOLFIRI and regorafenib combination therapy with dose escalation of irinotecan as fourth-line treatment for patients with metastatic colon cancer according to UGT1A1 genotyping. Irinotecan 68-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 25473295-1 2014 Here we report a case of metastatic colon cancer treated with 5-fluorouracil, leucovorin, and escalated doses of irinotecan (FOLFIRI) combined with regorafenib in the fourth-line setting after uridine diphosphate glucuronosyltransferase (UGT)1A1 genotyping analysis. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-245 25473295-1 2014 Here we report a case of metastatic colon cancer treated with 5-fluorouracil, leucovorin, and escalated doses of irinotecan (FOLFIRI) combined with regorafenib in the fourth-line setting after uridine diphosphate glucuronosyltransferase (UGT)1A1 genotyping analysis. FOLFIRI regimen 125-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-245 25473295-1 2014 Here we report a case of metastatic colon cancer treated with 5-fluorouracil, leucovorin, and escalated doses of irinotecan (FOLFIRI) combined with regorafenib in the fourth-line setting after uridine diphosphate glucuronosyltransferase (UGT)1A1 genotyping analysis. regorafenib 148-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 238-245 25473295-9 2014 FOLFIRI, with dose escalation of irinotecan according to UGT1A1 genotyping plus regorafenib appears to be a promising salvage therapy for patients with refractory metastatic colorectal cancer. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 25394051-4 2014 Atazanavir (ATV) inhibits UGT1A1, thus increasing unconjugated BR levels, a distinctive feature of this drug. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 25105254-2 2014 Bilirubin is a potent endogenous antioxidant, and the UGT1A1*28 polymorphism is the main genetic cause of variation in plasma bilirubin in Western Europe. Bilirubin 126-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 24910925-1 2014 AIM: To evaluate the impact of genetic polymorphisms in uridine 5"-glucuronosylytansferases UGT1A1 and UGT1A3 and iodothyronine-deiodinases types 1 and 2 on levothyroxine (T4 ; 3,5,3",5"-triiodo-L-thyronine) dose requirement for suppression of thyrotropin (TSH) secretion in patients with differentiated thyroid cancer (DTC). Uridine 56-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 25162819-5 2014 Prolonged raltegravir elimination likely reflects low neonatal UGT1A1 enzyme activity and enterohepatic recirculation. Raltegravir Potassium 10-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 25450281-1 2014 OBJECTIVE: The aim was to investigate the association between uridine diphosphate glucuronide transferase 1A1 (UGT1A1) gene promoter region polymorphism and irinotecan-related adverse effects and efficacy on recurrent and refractory small cell lung cancer (SCLC). Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-109 25450281-1 2014 OBJECTIVE: The aim was to investigate the association between uridine diphosphate glucuronide transferase 1A1 (UGT1A1) gene promoter region polymorphism and irinotecan-related adverse effects and efficacy on recurrent and refractory small cell lung cancer (SCLC). Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 25450281-4 2014 DNA was extracted from peripheral blood and direct sequencing method was employed to test UGT1A1FNx0128 polymorphism, thus analyzing the correlation between UGT1A1FNx0128 polymorphism and irinotecan-related side-effects and efficacy. Irinotecan 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 25204339-8 2014 Overall, the reversible, unconjugated hyperbilirubinemia associated with faldaprevir may predominantly result from inhibition of bilirubin conjugation by UGT1A1, with inhibition of hepatic uptake of bilirubin also potentially playing a role. faldaprevir 73-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 25204339-8 2014 Overall, the reversible, unconjugated hyperbilirubinemia associated with faldaprevir may predominantly result from inhibition of bilirubin conjugation by UGT1A1, with inhibition of hepatic uptake of bilirubin also potentially playing a role. Bilirubin 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 25204339-8 2014 Overall, the reversible, unconjugated hyperbilirubinemia associated with faldaprevir may predominantly result from inhibition of bilirubin conjugation by UGT1A1, with inhibition of hepatic uptake of bilirubin also potentially playing a role. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 25319636-0 2014 Crigler-Najjar syndrome type II in a Chinese boy resulting from three mutations in the bilirubin uridine 5"-diphosphate-glucuronosyltransferase (UGT1A1) gene and a family genetic analysis. Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 25493567-11 2014 Conclusion: We identified a UGT1A1 promoter variant (rs2003569) associated with simvastatin efficacy. Simvastatin 80-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 25319636-1 2014 BACKGROUND: The UGT1A1 gene encodes a responsible enzyme, UDP-glucuronosyltransferase1A1 (UGT1A1), for bilirubin metabolism. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 25319636-1 2014 BACKGROUND: The UGT1A1 gene encodes a responsible enzyme, UDP-glucuronosyltransferase1A1 (UGT1A1), for bilirubin metabolism. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-88 25319636-1 2014 BACKGROUND: The UGT1A1 gene encodes a responsible enzyme, UDP-glucuronosyltransferase1A1 (UGT1A1), for bilirubin metabolism. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 24709690-0 2014 The effect of the UGT1A1*28 allele on survival after irinotecan-based chemotherapy: a collaborative meta-analysis. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 25219630-7 2014 Capsaicin glucuronidation was significantly correlated with 3-O-glucuronidation of beta-estradiol (r = 0.637; p = 0.014) and with UGT1A1 protein levels (r = 0.616; p = 0.019) in a bank of individual HLMs (n = 14). Capsaicin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 25219630-9 2014 UGT1A1, 1A9 and 2B7 contributed 30.3, 6.0 and 49.0% of total glucuronidation of capsaicin in pHLM, respectively. Capsaicin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25219630-11 2014 CONCLUSION: Capsaicin was subjected to significant hepatic glucuronidation, wherein UGT1A1 and 2B7 were the main contributing enzymes. Capsaicin 12-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-98 24620945-2 2014 The high levels of bilirubin could be related to the co-inheritance of Gilbert syndrome determined either by mutations of the coding region or by variation in the (TA)n motifs of the promoter region of the bilirubin UGT1A1 gene. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 216-222 24620945-2 2014 The high levels of bilirubin could be related to the co-inheritance of Gilbert syndrome determined either by mutations of the coding region or by variation in the (TA)n motifs of the promoter region of the bilirubin UGT1A1 gene. Bilirubin 206-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 216-222 24620945-5 2014 METHODS: The promoter region (TA)n motifs of the bilirubin UGT1A1 gene were analyzed in 104 beta thalassemia individuals. Bilirubin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 24516079-1 2014 Hyperbilirubinaemia with or without jaundice is one of the side effects of atazanavir boosted with low-dose ritonavir (ATV/rit) related to the drug plasma levels, as a result of its metabolism by UGT1A1 - uridine diphosphate-glucuronosyl transferase. Atazanavir Sulfate 75-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 24516079-1 2014 Hyperbilirubinaemia with or without jaundice is one of the side effects of atazanavir boosted with low-dose ritonavir (ATV/rit) related to the drug plasma levels, as a result of its metabolism by UGT1A1 - uridine diphosphate-glucuronosyl transferase. Ritonavir 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 24516079-1 2014 Hyperbilirubinaemia with or without jaundice is one of the side effects of atazanavir boosted with low-dose ritonavir (ATV/rit) related to the drug plasma levels, as a result of its metabolism by UGT1A1 - uridine diphosphate-glucuronosyl transferase. Atazanavir Sulfate 119-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 24516079-2 2014 Genotyping for UGT1A1*28 before initiation of antiretroviral therapy containing atazanavir may aid in identifying individuals at risk of hyperbilirubinaemia. Atazanavir Sulfate 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 24516079-8 2014 Of the ATV/rit-treated patients, 14 were found to be carriers of the UGT1A1*28 variant (54%), and maximum serum bilirubin levels were significantly higher in the carrier population (4.71 vs. 2.69 mg/dL, p = 0.026). Atazanavir Sulfate 7-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 24516079-8 2014 Of the ATV/rit-treated patients, 14 were found to be carriers of the UGT1A1*28 variant (54%), and maximum serum bilirubin levels were significantly higher in the carrier population (4.71 vs. 2.69 mg/dL, p = 0.026). Ritonavir 11-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 24516079-8 2014 Of the ATV/rit-treated patients, 14 were found to be carriers of the UGT1A1*28 variant (54%), and maximum serum bilirubin levels were significantly higher in the carrier population (4.71 vs. 2.69 mg/dL, p = 0.026). Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 26692748-11 2014 In addition, if 7-hydroxymitragynine is to be taken with other drugs that are highly metabolized by UGT1A1, there is a possibility of drug-drug interaction to occur. 7-hydroxymitragynine 16-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 24709690-1 2014 To date, studies of irinotecan pharmacogenetics have mostly focused on the effect of the UGT1A1*28 allele on irinotecan-related toxicity. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 24709690-1 2014 To date, studies of irinotecan pharmacogenetics have mostly focused on the effect of the UGT1A1*28 allele on irinotecan-related toxicity. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 24709690-2 2014 However, the clinical utility of routine UGT1A1*28 genotyping to pre-emptively adjust irinotecan dosage is dependent upon whether UGT1A1*28 also affects patient survival following irinotecan therapy. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 24709690-2 2014 However, the clinical utility of routine UGT1A1*28 genotyping to pre-emptively adjust irinotecan dosage is dependent upon whether UGT1A1*28 also affects patient survival following irinotecan therapy. Irinotecan 180-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 24709690-4 2014 A systematic review and meta-analysis of both published and unpublished data were performed to summarize the available evidence of the relationship between the UGT1A1*28 allele and patient survival related to irinotecan therapy. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 25260839-0 2014 Influence of UGT1A1 gene methylation level in colorectal cancer cells on the sensitivity of the chemotherapy drug CPT-11. Irinotecan 114-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 25285015-0 2014 Clinical significance of UGT1A1 gene polymorphisms on irinotecan-based regimens as the treatment in metastatic colorectal cancer. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 25285015-1 2014 PURPOSE: The primary aim of this research was to investigate the association between uridine diphosphate glucuronosyltransferase (UGT)1A1 gene polymorphisms and the toxicities of irinotecan-based regimens in Chinese patients with metastatic colorectal cancer. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-137 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25210794-11 2014 However, HT29 cells became sensitized to the drug, and HSP90 client proteins were destabilized by ganetespib upon siRNA-mediated UGT1A knockdown. STA 9090 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-134 25210794-14 2014 We conclude that CRC cell-expressed UGT1A inactivates ganetespib and other resorcinolic Hsp90 inhibitors by glucuronidation, which renders the drugs unable to inhibit Hsp90 and thereby abrogates their biological activity. STA 9090 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-41 25210794-15 2014 UGT1A levels in tumor tissues may be a suitable predictive biomarker to stratify CRC patients for ganetespib treatment. STA 9090 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-5 25209391-0 2014 Glucose induces intestinal human UDP-glucuronosyltransferase (UGT) 1A1 to prevent neonatal hyperbilirubinemia. Glucose 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-70 25209391-2 2014 This study experimentally investigated whether additional glucose treatments induce the bilirubin-metabolizing enzyme--UDP-glucuronosyltransferase (UGT) 1A1--to prevent the onset of neonatal hyperbilirubinemia. Glucose 58-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-156 25209391-2 2014 This study experimentally investigated whether additional glucose treatments induce the bilirubin-metabolizing enzyme--UDP-glucuronosyltransferase (UGT) 1A1--to prevent the onset of neonatal hyperbilirubinemia. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-156 25209391-4 2014 In this study, UGT1A1 expression levels were determined in the liver and small intestine of neonatal hUGT1 mice that were orally treated with glucose. Glucose 142-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-106 25209391-5 2014 In the hUGT1 mice, glucose induced UGT1A1 in the small intestine, while it did not affect the expression of UGT1A1 in the liver. Glucose 19-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 7-12 25209391-6 2014 UGT1A1 was also induced in the human intestinal Caco-2 cells when the cells were cultured in the presence of glucose. Glucose 109-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25209391-8 2014 Adequate calorie intake would lead to the sufficient expression of UGT1A1 in the small intestine to reduce serum bilirubin levels. Bilirubin 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Glucuronides 4-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Estradiol 20-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Serotonin 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Propofol 54-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Zidovudine 64-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Zidovudine 92-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 24752421-6 2014 The glucuronides of estradiol, imipramine, serotonin, propofol, 3"-azido-3"-deoxythymidine (AZT) and morphine, selective substrates of human UGT1A1, 1A4, 1A6, 1A9 and 2B7 (AZT and morphine), respectively, were measured using LC-MS/MS. Morphine 101-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 25285015-14 2014 CONCLUSION: For irinotecan-based regimens in metastatic colorectal cancer, the UGT1A1*28 and UGT1A1*6 locus mutations can be regarded as predictors for irinotecan-associated severe delayed diarrhea, whereas no association between UGT1A1 gene polymorphisms and severe neutropenia was observed. Irinotecan 152-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 25044472-7 2014 UGT1A1, 1A3 and 1A10 showed activities on both 3"-OH and 7-OH, whereas UGT1A7, 1A8, 1A9, and 2B7 were only capable of catalyzing 3"-OH glucuronidation of calycosin. 3"-oh 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25044472-7 2014 UGT1A1, 1A3 and 1A10 showed activities on both 3"-OH and 7-OH, whereas UGT1A7, 1A8, 1A9, and 2B7 were only capable of catalyzing 3"-OH glucuronidation of calycosin. 7,3'-dihydroxy-4'-methoxyisoflavone 154-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25044472-9 2014 UGT1A1 showed the highest activity towards 7-OH glucuronidation (Clint, 224.34 muL/min/mg protein), which was comparable to its activity on 3"-OH glucuronidation (Clint, 203.82 muL/min/mg protein). 7-oh 43-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25044472-10 2014 Propofol (UGT1A9 inhibitor) produced a complete inhibition of 3"-glucuronide formation accompanied by an increase of 7-glucuronide in HLMs, while bilirubin (UGT1A1 inhibitor) only partially (~60%) inhibited the 7-OH glucuronidation. Bilirubin 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 25260839-1 2014 OBJECTIVE: To study the influence of the methylation level of UGT1A1 gene related to CPT-11 metabolic enzymes in colorectal cancer cells on the sensitivity of chemotherapy drugs. Irinotecan 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25260839-6 2014 CONCLUSIONS: The cytotoxicity of CPT-11 to colorectal cancer cells has a negative correlation with UGT1A1 expression, and positive correlation with CES2 and GUSB. Irinotecan 33-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 25260839-7 2014 The specific silencing UGT1A1 gene of siRNA could significantly increase the sensitivity of CPT-11 to the chemotherapy of colorectal cancer cells. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 25260839-8 2014 UGT1A1 methylation was an important factor affecting the chemosensitivity of CPT-11. Irinotecan 77-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24954688-4 2014 Kinetics studies were conducted to evaluate the K i values and mechanism(s) of the inhibition of CYP2E1, CYP3A4, UGT1A1 and UGT1A9 by p-cresol. 4-cresol 134-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 24954688-6 2014 p-Cresol was the most potent individual inhibitor, producing >50% inhibition of CYP2E1, CYP3A4, UGT1A1, UGT1A9 and UGT2B7 at a concentration of 100 muM. 4-cresol 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 24954688-9 2014 K i values for p-cresol inhibition of human liver microsomal CYP2E1, CYP3A4 and UGT1A1 ranged from 43 to 89 muM. 4-cresol 15-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 24635759-4 2014 Among 12 recombinant human UDP-glucuronosyltransferases (UGTs), UGT1A1, UGT1A8, UGT1A9, and UGT1A10 showed catalyzing activity toward leonurine glucuronidation. leonurine 134-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 25092941-3 2014 UGT1A1 is the rate-limiting enzyme in bilirubin"s metabolism. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 25054039-0 2014 Clinical usefulness of testing for UDP glucuronosyltransferase 1 family, polypeptide A1 polymorphism prior to the inititation of irinotecan-based chemotherapy. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-87 25054039-1 2014 An association between UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) polymorphisms and irinotecan-induced neutropenia has been previously reported. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-75 25054039-1 2014 An association between UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) polymorphisms and irinotecan-induced neutropenia has been previously reported. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24635759-6 2014 Both chemical inhibition study and correlation study demonstrated that leonurine glucuronidation activities in HLMs had significant relationship with UGT1A1 activities. leonurine 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 24635759-8 2014 UGT1A1 was principal enzyme that responsible for leonurine glucuronidation in human liver and intestine microsomes. leonurine 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24650397-10 2014 CONCLUSION: One-half of the infants with BMJ were homozygous UGT1A1*6 and exhibited a serum bilirubin concentration significantly greater than other genotypes. Bilirubin 92-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 24928535-6 2014 The up-regulation of MDR1 and UGT1A1 by anticancer agents was further enhanced in the presence of CITCO. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 98-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 24462762-3 2014 The study group received irinotecan dose escalation based on UGT1A1 genotyping whereas the control group did not. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 24462762-5 2014 The clinical response rate of patients with mCRC treated with FOLFIRI plus bevacizumab under UGT1A1 genotyping and irinotecan dose escalation was significantly better than that of those without these prospective tests and dose escalation (P = 0.028). FOLFIRI regimen 62-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24462762-8 2014 Patients with mCRC undergoing UGT1A1 genotyping may receive escalated doses of irinotecan to obtain a better clinical response/outcome with comparable toxicities. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 24865931-4 2014 Genetic polymorphisms of the UGT1A1 promoter, specifically the -3279 T G phenobarbital responsive enhancer module (rs4124874) and (TA)7 dinucleotide repeat (rs8175347) as well as the coding region variants (rs2306283 and rs4149056) of the OATP2 gene were significantly higher among the cases than the controls. Phenobarbital 73-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 24865931-7 2014 This study demonstrates that UGT1A1 and OATP2 polymorphisms were associated with altered bilirubin metabolism and could be genetic risk factors for neonatal hyperbilirubinemia. Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 24785582-1 2014 BACKGROUND: Gilbert"s syndrome is one of the most common metabolic syndromes in the human population characterised by mild unconjugated hyperbilirubinemia resulting from reduced activity of the bilirubin conjugating enzyme UDP-glucuronosyltransferase (UGT1A1). Bilirubin 141-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 252-258 24958824-0 2014 Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-1 2014 PURPOSE: The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. Irinotecan 178-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24958824-14 2014 CONCLUSION: The UGT1A1*28 genotype can be used to individualize dosing of irinotecan. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 24837423-3 2014 The hypothesis of a system is supported by (i) coordinate regulation of subsets of these enzyme families and transporters by transcription factors including the AhR, and (ii) feedback loops between endobiotic AhR agonists and substrates of major catabolic target genes/proteins; for example, 6-formylindolo[3,2-b]carbazole as substrate of CYP1A1, and bilirubin, as substrate of UGT1A1. 6-formylindolo(3,2-b)carbazole 292-322 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 378-384 24837423-3 2014 The hypothesis of a system is supported by (i) coordinate regulation of subsets of these enzyme families and transporters by transcription factors including the AhR, and (ii) feedback loops between endobiotic AhR agonists and substrates of major catabolic target genes/proteins; for example, 6-formylindolo[3,2-b]carbazole as substrate of CYP1A1, and bilirubin, as substrate of UGT1A1. Bilirubin 351-360 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 378-384 24650397-2 2014 STUDY DESIGN: UGT1A1 gene allelic variation was analyzed in 170 Japanese infants with BMJ with polymerase chain reaction-direct sequencing, and their genotypes compared with serum bilirubin concentrations. Bilirubin 180-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 24650397-12 2014 Previous finding have demonstrated that 5beta-pregnane-3alpha,20beta-diol present in breast milk inhibits p.G71R-UGT1A1 bilirubin glucuronidation activity. 5beta-pregnane-3alpha 40-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 24650397-12 2014 Previous finding have demonstrated that 5beta-pregnane-3alpha,20beta-diol present in breast milk inhibits p.G71R-UGT1A1 bilirubin glucuronidation activity. 20beta-diol 62-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 24650397-12 2014 Previous finding have demonstrated that 5beta-pregnane-3alpha,20beta-diol present in breast milk inhibits p.G71R-UGT1A1 bilirubin glucuronidation activity. Bilirubin 120-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 24901842-2 2014 The UGT1A1*28 polymorphism (TA repeats in the promoter region) is a major determinant of bilirubin levels and recent evidence suggests that raised adiposity may also be a contributing factor. Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 24857157-7 2014 Subsequently, the mRNA levels of AHR-targeting drug-metabolizing enzymes (CYP1A1, UGT1A1, and ABCG 2) and the protein level of CYP1A1 in HepG2 cells were shown to be increased by 6-shogaol. shogaol 179-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 24901842-3 2014 We aimed to study the interaction between UGT1A1 polymorphism, hematological and anthropometric variables with total bilirubin levels in young individuals. Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 24901842-12 2014 UGT1A1 polymorphism and body fat percentage were the main factors affecting bilirubin levels within obese patients (linear regression analysis). Bilirubin 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24901842-13 2014 CONCLUSION: In obese children and adolescents, body fat composition and UGT1A1 polymorphism are independent determinants of total bilirubin levels. Bilirubin 130-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 24977443-0 2014 [Interest of UGT1A1 genotyping within digestive cancers treatment by irinotecan]. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24977443-2 2014 Its anticancer activity results from its bioactivation into SN-38 metabolite, which is cleared through glucuronidation by the hepatic enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-188 24977443-2 2014 Its anticancer activity results from its bioactivation into SN-38 metabolite, which is cleared through glucuronidation by the hepatic enzyme uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 24977443-9 2014 The existence of a French laboratories network performing this test in clinical routine makes it possible to generalize UGT1A1 deficiency screening, so as to guarantee equal access to safe treatment and optimized irinorecan-based therapy for the many patients receiving irinotecan-based therapy in advanced colorectal cancer. irinorecan 213-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 24977443-9 2014 The existence of a French laboratories network performing this test in clinical routine makes it possible to generalize UGT1A1 deficiency screening, so as to guarantee equal access to safe treatment and optimized irinorecan-based therapy for the many patients receiving irinotecan-based therapy in advanced colorectal cancer. Irinotecan 270-280 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 24777631-6 2014 Rifampicin is also an inducer of the uridine diphosphate glucuronosyltransferase (UGT) 1A1 enzymes and interferes with drugs, such as integrase inhibitors, that are metabolized by this metabolic pathway. Rifampin 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-90 25141892-0 2014 UGT1A1*28 polymorphisms: a potential pharmacological biomarker of irinotecan-based chemotherapies in colorectal cancer. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24932285-0 2014 Association between the low-dose irinotecan regimen-induced occurrence of grade 4 neutropenia and genetic variants of UGT1A1 in patients with gynecological cancers. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-171 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-171 24932285-1 2014 The occurrence of severe neutropenia during treatment with irinotecan (CPT-11) is associated with the *6 and *28 alleles of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 173-179 24932285-6 2014 The association between the absolute neutrophil count (ANC) nadir values, the total dose of CPT-11 and the genotypes of UGT1A1 or ABCG2 was studied. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 24932285-12 2014 In conclusion, the UGT1A1*6/*28 and *6/*6 genotypes were found to be associated with the occurrence of severe neutropenia in the low-dose CPT-11 regimen for gynecological cancers. Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 24932285-13 2014 This finding indicates that the determination of UGT1A1 variants may be as useful in CPT-11 chemotherapy for gynecological conditions as it is in colorectal and lung cancer patients treated with this drug. Irinotecan 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 24453052-10 2014 A comprehensive study of the influence of UGT1A1 polymorphisms on the risk of irinotecan-induced toxicity is necessary for optimal use of irinotecan treatment. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 24453052-10 2014 A comprehensive study of the influence of UGT1A1 polymorphisms on the risk of irinotecan-induced toxicity is necessary for optimal use of irinotecan treatment. Irinotecan 138-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 24375494-0 2014 Analysis of flavonoids regulating the expression of UGT1A1 via xenobiotic receptors in intestinal epithelial cells. Flavonoids 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 24834123-2 2014 Our purpose was to study the association between UGT1A1*28, UGT1A7*2, and UGT1A7*3 polymorphisms and irinotecan toxicity in Greek patients receiving low-dose weekly irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 24375494-2 2014 A screening assay using real-time PCR showed that baicalein and 3-hydroxyflavone induced human UGT1A1 mRNA expression in LS180 cells. baicalein 50-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 24375494-2 2014 A screening assay using real-time PCR showed that baicalein and 3-hydroxyflavone induced human UGT1A1 mRNA expression in LS180 cells. 3-hydroxyflavone 64-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 24375494-4 2014 The results indicated that baicalein and 3-hydroxyflavone increased the transcriptional activity of UGT1A1 via AhR and PXR, respectively. baicalein 27-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 24375494-4 2014 The results indicated that baicalein and 3-hydroxyflavone increased the transcriptional activity of UGT1A1 via AhR and PXR, respectively. 3-hydroxyflavone 41-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 24375494-7 2014 These results elucidate the molecular mechanism of flavonoid-induced UGT1A1 gene expression via xenobiotic receptors in the intestines. Flavonoids 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 24598415-6 2014 Regulation of phase II rather than phase I metabolic enzymes was implicated mechanistically: raloxifene and DMA were observed to upregulate sulfotransferase (SULT 1E1) and glucuronidase (UGT 1A1). Raloxifene Hydrochloride 93-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-194 24598415-6 2014 Regulation of phase II rather than phase I metabolic enzymes was implicated mechanistically: raloxifene and DMA were observed to upregulate sulfotransferase (SULT 1E1) and glucuronidase (UGT 1A1). desmethylarzoxifene 108-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-194 23981182-3 2014 METHODS: We have investigated the UGT1A1 polymorphism frequency and its relation to severity of hyperbilirubinemia and jaundice duration among 220 exclusively breastfed term newborns; 57 of them with non-physiologic hyperbilirubinemia (NH), and 163 with BMJ, and in 187 healthy controls. (12R)-12-methyltetradecanoic acid 254-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 24382596-0 2014 Pharmacokinetics, safety, and efficacy of FOLFIRI plus bevacizumab in Japanese colorectal cancer patients with UGT1A1 gene polymorphisms. FOLFIRI regimen 42-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 24382596-1 2014 Previous reports of the influence of UGT1A1 gene polymorphisms on the pharmacokinetics of irinotecan metabolism have not assessed Asian patients treated with FOLFIRI plus bevacizumab for advanced and recurrent colorectal cancer. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 24627218-5 2014 In addition, inhibition of UDP-glucuronosyltransferase 1A1 (UGT1A1) variants by lenalidomide was also assessed. Lenalidomide 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-58 24519753-0 2014 Associations between UGT1A1*6 or UGT1A1*6/*28 polymorphisms and irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 64-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 24519753-2 2014 Many studies have demonstrated that patients bearing UGT1A1*28 have a higher risk of severe neutropenia on toxicity of irinotecan. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 24553666-5 2014 Recombinant UGT enzymes (UGT1A1, 1A3, 1A4, 1A6, 1A9, 2B7, 2B10, 2B15) were screened for their activity towards ethanol, and kinetic data were then established for all enzymes. Ethanol 111-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 24627218-5 2014 In addition, inhibition of UDP-glucuronosyltransferase 1A1 (UGT1A1) variants by lenalidomide was also assessed. Lenalidomide 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 24519753-4 2014 Some researches reported that UGT1A1*28 and/or UGT1A1*6 could predict irinotecan-induced toxicities in Asian populations, but controversial conclusions still remained. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 24519753-4 2014 Some researches reported that UGT1A1*28 and/or UGT1A1*6 could predict irinotecan-induced toxicities in Asian populations, but controversial conclusions still remained. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-109 24519753-5 2014 This study aims to investigate the association between UGT1A1 gene polymorphisms *6, *6/*28 and irinotecan-related neutropenia in Asian cancer patients receiving irinotecan regimen chemotherapy. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24519753-5 2014 This study aims to investigate the association between UGT1A1 gene polymorphisms *6, *6/*28 and irinotecan-related neutropenia in Asian cancer patients receiving irinotecan regimen chemotherapy. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24519753-6 2014 EXPERIMENTAL DESIGN: Meta-analyses were done to assess the relationship between UGT1A1*6 or UGT1A1*6/*28 and irinotecan-induced neutropenia. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 24519753-6 2014 EXPERIMENTAL DESIGN: Meta-analyses were done to assess the relationship between UGT1A1*6 or UGT1A1*6/*28 and irinotecan-induced neutropenia. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 24519753-9 2014 CONCLUSIONS: In conclusion, the UGT1A1*6 and UGT1A1*6/*28 genotypes were associated with an increased risk of irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24519753-9 2014 CONCLUSIONS: In conclusion, the UGT1A1*6 and UGT1A1*6/*28 genotypes were associated with an increased risk of irinotecan-induced neutropenia in Asian cancer patients. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 179-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-109 23605141-1 2014 BACKGROUND: It was recently reported that genetic polymorphisms of UDP glucuronyltransferase-1 polypeptide A1 (UGT1A1), a glucuronidation enzyme, were associated with irinotecan (CPT-11) metabolism. Irinotecan 179-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 23605141-2 2014 The active metabolite of CPT-11, 7-ethyl-10-hydroxycamptothecin (SN-38) was glucuronidated (SN-38G) by UGT1A1. cpt-11, 7-ethyl-10-hydroxycamptothecin 25-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23605141-2 2014 The active metabolite of CPT-11, 7-ethyl-10-hydroxycamptothecin (SN-38) was glucuronidated (SN-38G) by UGT1A1. Irinotecan 65-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23605141-2 2014 The active metabolite of CPT-11, 7-ethyl-10-hydroxycamptothecin (SN-38) was glucuronidated (SN-38G) by UGT1A1. Irinotecan 92-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23605141-4 2014 Several studies have suggested that the dose of CPT-11 should be decreased in patients homozygous for UGT1A1*6 or UGT1A1*28, or double heterozygotes (*6/*28). Irinotecan 48-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 23605141-4 2014 Several studies have suggested that the dose of CPT-11 should be decreased in patients homozygous for UGT1A1*6 or UGT1A1*28, or double heterozygotes (*6/*28). Irinotecan 48-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 13-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 13-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 13-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-7 2014 RESULTS: The SN-38G/SN-38 concentration ratio was lower in patients who were homozygous for UGT1A1*6, heterozygous for UGT1A1*6 or UGT1A1*28, or were double heterozygotes compared with patients with wild-type genes. Irinotecan 13-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 30-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 30-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 30-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23605141-8 2014 The relative decreases in the SN-38G/SN-38 concentration ratio in patients homozygous for UGT1A1*6 and in double heterozygotes were greater than in patients heterozygous for UGT1A1*6 or UGT1A1*28. Irinotecan 30-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 23609856-0 2014 Association between severe toxicity of nilotinib and UGT1A1 polymorphisms in Japanese patients with chronic myelogenous leukemia. nilotinib 39-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 23609856-2 2014 The UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism UGT1A1*28 (*28)/*28 has been linked to an increased risk of hyperbilirubinemia in patients with CML who receive nilotinib. nilotinib 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-35 23609856-2 2014 The UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism UGT1A1*28 (*28)/*28 has been linked to an increased risk of hyperbilirubinemia in patients with CML who receive nilotinib. nilotinib 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 23609856-2 2014 The UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism UGT1A1*28 (*28)/*28 has been linked to an increased risk of hyperbilirubinemia in patients with CML who receive nilotinib. nilotinib 170-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 23609856-3 2014 Beside *28, UGT1A1*6 (*6) is another important variant allele in Japanese patients because it is associated with adverse events of irinotecan, metabolized by UGT1A1. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23609856-3 2014 Beside *28, UGT1A1*6 (*6) is another important variant allele in Japanese patients because it is associated with adverse events of irinotecan, metabolized by UGT1A1. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 23609856-4 2014 We retrospectively investigated the association between severe toxicity of nilotinib and UGT1A1 polymorphisms (*6 and*28) in Japanese patients with CML. nilotinib 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 23609856-9 2014 CONCLUSION: These findings suggest that UGT1A1 polymorphisms are important determinants of severe toxicity of nilotinib in Japanese patients. nilotinib 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 24525562-0 2014 Simultaneous determination of bilirubin and its glucuronides in liver microsomes and recombinant UGT1A1 enzyme incubation systems by HPLC method and its application to bilirubin glucuronidation studies. Bilirubin 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 24525562-1 2014 Bilirubin, an important endogenous substances and liver function index in humans, is primarily eliminated via UGT1A1-catalyzed glucuronidation. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 24525562-8 2014 Furthermore, we established stable, reliable in vitro incubation systems and optimized the incubation conditions to characterize the kinetics of bilirubin glucuronidation by RLM, HLM and UGT1A1, respectively. Bilirubin 145-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 24525562-10 2014 Bilirubin glucuronidation obeyed the Hill equation by RLM and the Michaelis-Menten equation by HLM and UGT1A1 in the range of substrate concentration selected, respectively. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 24525562-11 2014 In addition, the relative proportions between BDG and BMGs were in connection with enzyme sources (e.g. RLM, HLM and UGT1A1) and bilirubin concentration. O(6)-n-butyldeoxyguanosine 46-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 23529007-0 2014 Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 23529007-1 2014 A meta-analysis in Caucasians was conducted to investigate the possible association of uridine diphosphate glucuronosyltransferase (UGT) 1A1 gene polymorphisms with irinotecan (IRI)-induced neutropenia and diarrhoea in colorectal cancer (CRC). Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-140 23529007-8 2014 In subgroup meta-analysis, the higher incidence of diarrhoea in UGT1A1*28/*28 patients was limited to studies where when IRI was given at higher doses (OR=2.37, 95% CI: 1.39-4.04; P=0.002) or combined with 5-fluorouracil (FU or analogue) (OR=1.78, 95% CI: 1.16-2.75; P=0.009). Fluorouracil 206-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 23529007-10 2014 IRI-combined 5-FU (or analogue) and a high-dose IRI therapy enhance IRI-induced diarrhoea among patients bearing the UGT1A1*28 allele. Fluorouracil 13-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 24407487-4 2014 The (GT)n and (TA)n dinucleotide variations in heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) gene promoters were determined by fragment analysis. Dinucleoside Phosphates 20-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 24557078-7 2014 By multivariate analysis, higher peak on-treatment bilirubin levels were found to be associated with the UGT1A1 rs887829 T allele (P=6.4x10(-12)), higher baseline hemoglobin levels (P=4.9x10(-13)), higher baseline bilirubin levels (P=6.7x10(-12)), and slower plasma atazanavir clearance (P=8.6x10(-11)). Bilirubin 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 24557078-11 2014 CONCLUSION: Atazanavir-associated hyperbilirubinemia is best predicted by considering UGT1A1 genotype, baseline bilirubin level, and baseline hemoglobin level in combination. Atazanavir Sulfate 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 24557078-11 2014 CONCLUSION: Atazanavir-associated hyperbilirubinemia is best predicted by considering UGT1A1 genotype, baseline bilirubin level, and baseline hemoglobin level in combination. Bilirubin 39-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 24690955-2 2014 Bilirubin is produced by heme oxygenase-1 (HO-1), catalyzed by UDP-glucuronosyltransferase (UGT1A1), and has potential as an antioxidant. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 24690955-8 2014 Post-transplant 1-year bilirubin level was higher in 6/7 or 7/7 carriers compared with 6/6 homozygotes in terms of the UGT1A1*28 polymorphism (6/6 vs. 6/7 vs. 7/7: 0.71 +- 0.27 vs. 1.06 +- 0.36 vs. 1.10 +- 0.45 mg/dL, P<0.001). Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 24690955-12 2014 CONCLUSIONS: The UGT1A1*28 polymorphism is associated with changes in serum bilirubin and with graft outcome after kidney transplantation. Bilirubin 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 24066723-2 2014 Nilotinib, a tyrosine kinase inhibitor, could potently inhibit SN-38 glucuronidation mainly catalyzed by UDP-glucuronosyltransferase (UGT) 1A1. nilotinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-142 24066723-2 2014 Nilotinib, a tyrosine kinase inhibitor, could potently inhibit SN-38 glucuronidation mainly catalyzed by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-142 24066723-3 2014 This study was designed to investigate whether nilotinib can be used as a selective inhibitor of UGT1A1 in human liver. nilotinib 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 24066723-5 2014 Assays with recombinant UGTs indicated that nilotinib could strongly inhibit the activity of UGT1A1 and decreased the activity of extra-hepatic UGT1A7 to a much lesser extent. nilotinib 44-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24066723-6 2014 The inhibition on 4-methylumbelliferone (4Mu) glucuronidation by recombinant UGT1A1 obeyed competitive inhibition mechanism, with a kinetic constants (Ki) value of 0.17 muM. Hymecromone 18-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24066723-7 2014 Assays with human liver microsomes (HLM) demonstrated that nilotinib could selectively inhibit estradiol-3-O-glucuronidation (E2-3-O-glucuronidation), a probe reaction of UGT1A1. nilotinib 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 24066723-11 2014 Given that UGT1A7 is an extra-hepatic enzyme, this study indicates that nilotinib can be used as a selective inhibitor of UGT1A1 in human liver. nilotinib 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 23996546-5 2014 The bergenin glucuronosyltransferase activities in HLMs and UGT1A1 were inhibited by phenylbutazone, estradiol and bilirubin. Phenylbutazone 85-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 23996546-5 2014 The bergenin glucuronosyltransferase activities in HLMs and UGT1A1 were inhibited by phenylbutazone, estradiol and bilirubin. Estradiol 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 23996546-5 2014 The bergenin glucuronosyltransferase activities in HLMs and UGT1A1 were inhibited by phenylbutazone, estradiol and bilirubin. Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 24448639-0 2014 UGT1A1*6 polymorphisms are correlated with irinotecan-induced toxicity: a system review and meta-analysis in Asians. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 5-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-1 2014 AIM: 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of irinotecan, is mainly eliminated hepatically through glucuronidation by UGT1A1 and UGT1A9 enzymes. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 24897286-2 2014 This study comprehensively investigates the effects of UGT1A1 and UGT1A9 genetic polymorphism on SN-38 glucuronidation activity. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24897286-5 2014 The number of UGT1A1 reduced function alleles was associated with decreased SN-38G formation rates and UGT1A protein levels. Irinotecan 76-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 24897286-8 2014 CONCLUSION: UGT1A1 genetic polymorphisms have a more important function in human liver SN-38 glucuronidation activity than UGT1A9. Irinotecan 87-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 24696866-7 2014 Elvitegravir undergoes extensive primary metabolism by hepatic and intestinal cytochrome P450 3A (CYP3A) and secondary metabolism by UDP-glucuronosyltransferase 1-1 and 1-3 (UGT1A1/3). elvitegravir 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 24407487-7 2014 Serum bilirubin levels were significantly lower in patients with CD within all UGT1A1*28 genotypes (P < 0.05). Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 24407487-11 2014 UGT1A1*28 allele homozygosity, responsible for higher bilirubin levels, seems to be an important modifier of CD manifestation. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24318863-5 2014 Target genes were XPD-751, GSTP-1-105, XRCC1-399 for oxaliplatin, UGT1A1 for irinotecan. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 24399855-6 2014 For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4"-glucuronide (ral-4"-gluc; P < 0.01). Raloxifene Hydrochloride 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 24399855-6 2014 For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4"-glucuronide (ral-4"-gluc; P < 0.01). Raloxifene 6-Glucuronide 96-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 23886114-3 2014 Genome-wide association studies have shown significant association of OATP1B1 and UGT1A1 with elevations of unconjugated bilirubin, and OATP1B1 inhibition data correlated with clinical unconjugated hyperbilirubinemia for several compounds. Bilirubin 121-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 24399855-6 2014 For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4"-glucuronide (ral-4"-gluc; P < 0.01). ral-6-gluc 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 24399855-6 2014 For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4"-glucuronide (ral-4"-gluc; P < 0.01). Raloxifene 4'-Glucuronide 151-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 24399855-6 2014 For UGT1A1, this repression correlated with significantly reduced metabolism of raloxifene into raloxifene-6-glucuronide (ral-6-gluc; P < 0.01) and raloxifene-4"-glucuronide (ral-4"-gluc; P < 0.01). ral-4"-gluc 178-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 24308720-0 2014 The association of UGT1A1*6 and UGT1A1*28 with irinotecan-induced neutropenia in Asians: a meta-analysis. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 24587300-0 2014 Serum bilirubin concentration in healthy adult North-Europeans is strictly controlled by the UGT1A1 TA-repeat variants. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24587300-1 2014 The major enzyme responsible for the glucuronidation of bilirubin is the uridine 5"-diphosphoglucose glucuronosyltransferase A1 (UGT1A1) enzyme, and genetic variation in the UGT1A1 gene is reported to influence the bilirubin concentration in the blood. Bilirubin 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 24587300-1 2014 The major enzyme responsible for the glucuronidation of bilirubin is the uridine 5"-diphosphoglucose glucuronosyltransferase A1 (UGT1A1) enzyme, and genetic variation in the UGT1A1 gene is reported to influence the bilirubin concentration in the blood. Bilirubin 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 24587300-1 2014 The major enzyme responsible for the glucuronidation of bilirubin is the uridine 5"-diphosphoglucose glucuronosyltransferase A1 (UGT1A1) enzyme, and genetic variation in the UGT1A1 gene is reported to influence the bilirubin concentration in the blood. uridine 5"- 73-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 24587300-1 2014 The major enzyme responsible for the glucuronidation of bilirubin is the uridine 5"-diphosphoglucose glucuronosyltransferase A1 (UGT1A1) enzyme, and genetic variation in the UGT1A1 gene is reported to influence the bilirubin concentration in the blood. Bilirubin 215-224 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 24587300-1 2014 The major enzyme responsible for the glucuronidation of bilirubin is the uridine 5"-diphosphoglucose glucuronosyltransferase A1 (UGT1A1) enzyme, and genetic variation in the UGT1A1 gene is reported to influence the bilirubin concentration in the blood. Bilirubin 215-224 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 24587300-5 2014 We found significant odds ratios for high bilirubin level for all the selected UGT1A1 variants. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 24587300-6 2014 However, in stepwise multivariate logistic regression analysis of all genetic variants together with age, sex, country of origin and fasting time, the repeat variants of UGT1A1 TA6>TA7 and SLCO1B3 rs2117032 T>C were the only variants significantly associated with higher bilirubin concentrations. Bilirubin 277-286 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 24587300-8 2014 Among individuals heterozygous for the TA7-repeat, a low frequent UGT1A1-diplotype harboring the rs7564935 G-variant was associated with higher bilirubin levels. Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 24308720-0 2014 The association of UGT1A1*6 and UGT1A1*28 with irinotecan-induced neutropenia in Asians: a meta-analysis. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24308720-1 2014 BACKGROUND: The UGT1A1*28 polymorphism is known as a biomarker of irinotecan-induced neutropenia in Caucasians. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 24308720-5 2014 CONCLUSIONS: In Asians, a combination test of UGT1A1*6 and UGT1A1*28 might be a potential biomarker of irinotecan-induced neutropenia, an observation that will need additional trials for confirmation. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 24308720-5 2014 CONCLUSIONS: In Asians, a combination test of UGT1A1*6 and UGT1A1*28 might be a potential biomarker of irinotecan-induced neutropenia, an observation that will need additional trials for confirmation. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 23888985-7 2014 Glucuronide capacity was present for the substrates 17beta-estradiol (estradiol-3-glucuronide, 2.9 +- 0.2 nmol/mg protein/min) and 4-methylumbelliferone (31.3 +- 3.3 nmol/mg protein/min), assuming that cats have functional homologue enzymes to at least the human UGT1A1 and probably other UGT1A isozymes. Glucuronides 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 263-269 24439671-0 2014 Regulation and expression of aberrant methylation on irinotecan metabolic genes CES2, UGT1A1 and GUSB in the in-vitro cultured colorectal cancer cells. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 24439671-8 2014 CONCLUSION: Methylation in UGT1A1 gene expression silencing as an important mechanism; methylation could provide an effective target for methylation regulation intervening in the treatment of CPT-11. Irinotecan 192-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 24243081-5 2014 Application of our model revealed that an absolute value or a steady state increase in bilirubin falling below 3.8Phi micromol/L (where Phi is a correction factor, =1 for UGT1A1 wild type and 1 for UGT1A1 variants) could be used to predict suboptimal atazanavir exposure and treatment failure. Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 24243081-5 2014 Application of our model revealed that an absolute value or a steady state increase in bilirubin falling below 3.8Phi micromol/L (where Phi is a correction factor, =1 for UGT1A1 wild type and 1 for UGT1A1 variants) could be used to predict suboptimal atazanavir exposure and treatment failure. Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 24243081-5 2014 Application of our model revealed that an absolute value or a steady state increase in bilirubin falling below 3.8Phi micromol/L (where Phi is a correction factor, =1 for UGT1A1 wild type and 1 for UGT1A1 variants) could be used to predict suboptimal atazanavir exposure and treatment failure. Atazanavir Sulfate 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 24243081-5 2014 Application of our model revealed that an absolute value or a steady state increase in bilirubin falling below 3.8Phi micromol/L (where Phi is a correction factor, =1 for UGT1A1 wild type and 1 for UGT1A1 variants) could be used to predict suboptimal atazanavir exposure and treatment failure. Atazanavir Sulfate 252-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 24243081-6 2014 Thus, we have successfully established a new mathematical approach for pharmacodynamic-pharmacokinetic modelling of the interaction between atazanavir and bilirubin, as it relates to genetic variants of UGT1A1. Atazanavir Sulfate 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-209 24243081-6 2014 Thus, we have successfully established a new mathematical approach for pharmacodynamic-pharmacokinetic modelling of the interaction between atazanavir and bilirubin, as it relates to genetic variants of UGT1A1. Bilirubin 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-209 23888985-7 2014 Glucuronide capacity was present for the substrates 17beta-estradiol (estradiol-3-glucuronide, 2.9 +- 0.2 nmol/mg protein/min) and 4-methylumbelliferone (31.3 +- 3.3 nmol/mg protein/min), assuming that cats have functional homologue enzymes to at least the human UGT1A1 and probably other UGT1A isozymes. Estradiol 52-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 263-269 23888985-7 2014 Glucuronide capacity was present for the substrates 17beta-estradiol (estradiol-3-glucuronide, 2.9 +- 0.2 nmol/mg protein/min) and 4-methylumbelliferone (31.3 +- 3.3 nmol/mg protein/min), assuming that cats have functional homologue enzymes to at least the human UGT1A1 and probably other UGT1A isozymes. estradiol-3-glucuronide 70-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 263-269 23888985-7 2014 Glucuronide capacity was present for the substrates 17beta-estradiol (estradiol-3-glucuronide, 2.9 +- 0.2 nmol/mg protein/min) and 4-methylumbelliferone (31.3 +- 3.3 nmol/mg protein/min), assuming that cats have functional homologue enzymes to at least the human UGT1A1 and probably other UGT1A isozymes. Hymecromone 131-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 263-269 24815493-0 2014 Clinical observations on associations between the UGT1A1 genotype and severe toxicity of irinotecan. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 24815493-2 2014 UDP glucuronosyltransferase1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38 but the relationship between UGT1A1 and severe toxicity remains unclear. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 24815493-10 2014 CONCLUSIONS: Our study support the conclusion that patients with a UGT1A1*28 allele (s) will suffer an increased risk of severe irinotecan-induced diarrhea, whether with mid-or low-dosage. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 24815493-12 2014 Higher serum total bilirubin is an indication that patients UGT1A1 genotype is not wild-type, with significance for clinic usage of CPT-11. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 24815493-12 2014 Higher serum total bilirubin is an indication that patients UGT1A1 genotype is not wild-type, with significance for clinic usage of CPT-11. Irinotecan 132-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 25967674-0 2014 Prevalence of the UGT1A1*6 (c.211G>A) Polymorphism and Prediction of Irinotecan Toxicity in Iranian Populations of Different Ethnicities. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 25967674-1 2014 BACKGROUND: Pharmacogenetic studies on irinotecan treatment in patients with metastatic colorectal cancer have indicated that genetic polymorphisms in UGT1A1*6 can lead to decreased enzyme activity and accumulation of the toxic metabolite SN-38. Irinotecan 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 25967674-1 2014 BACKGROUND: Pharmacogenetic studies on irinotecan treatment in patients with metastatic colorectal cancer have indicated that genetic polymorphisms in UGT1A1*6 can lead to decreased enzyme activity and accumulation of the toxic metabolite SN-38. Irinotecan 239-244 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 25967674-9 2014 CONCLUSION: The identification of the UGT1A1*6 alleles is necessary among the different Iranian ethnic groups before irinotecan therapy, suggesting that genotyping would be helpful for clinicians to optimize chemotherapy or identify individuals at risk of adverse drug reactions before clinical trials. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 24104197-4 2014 The polymorphisms evaluated included those in genes encoding putative acetaminophen-metabolizing enzymes (UGT1A1, UGT1A6, UGT1A9, UGT2B15, SULT1A1, CYP2E1, and CYP3A5) as well as CD44 and BHMT1. Acetaminophen 70-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Bilirubin 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-60 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Bilirubin 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-60 25016708-1 2014 BACKGROUND: Uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is a key conjugating enzyme of bilirubin and the anti-tumor medication irinotecan. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 25016708-2 2014 Comprehensive analysis of UGT1A1 gene polymorphisms may provide benefit by predicting pharmacokinetics and outcomes of treatment with irinotecan and certain antiviral medications. Irinotecan 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23965644-6 2014 We demonstrate that UGT1A1 and UGT1A3 are mainly responsible for pterostilbene glucuronidation although UGT1A8, UGT1A9 and UGT1A10 also had detectable activity. pterostilbene 65-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23965644-7 2014 Intriguingly, UGT1A1 exhibits the highest activity against both resveratrol and pterostilbene despite altered hydroxyl group specificity. Resveratrol 64-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 23965644-7 2014 Intriguingly, UGT1A1 exhibits the highest activity against both resveratrol and pterostilbene despite altered hydroxyl group specificity. pterostilbene 80-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 24055854-6 2014 17beta-Estradiol was used as the marker substrate to determine UGT1A1 activities. Estradiol 0-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 23965644-7 2014 Intriguingly, UGT1A1 exhibits the highest activity against both resveratrol and pterostilbene despite altered hydroxyl group specificity. Hydroxyl Radical 110-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 24025985-6 2014 UGT1A1, -1A6, -1A7, -1A8, -1A9 and -1A10 participated in the formation of 7-O-G, while the formation of 8-O-G was catalyzed selectively by UGT1A6 and UGT1A9. 7-o-g 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24898899-0 2014 Influence of UGT1A1 6, 27, and 28 polymorphisms on nilotinib-induced hyperbilirubinemia in Japanese patients with chronic myeloid leukemia. nilotinib 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24898899-1 2014 Nilotinib potently inhibits human uridine diphosphate-glucuronosyltransferase (UGT1A1) activity, causing hyperbilirubinemia. nilotinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 24898899-4 2014 The median time to elevation of bilirubin levels in UGT1A1 poor metabolizers was 2.0 weeks (hazard ratio, 6.11). Bilirubin 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 24898899-5 2014 The median time to reduction in nilotinib dose in UGT1A1 poor metabolizers was 4.0 weeks (hazard ratio, 7.52; p = 0.002). nilotinib 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 24898899-6 2014 Consequently, in the maintenance phase 3 months following the initiation of nilotinib therapy, the median daily dose and C0 of nilotinib were 350 mg/day and 372 ng/mL, respectively, in UGT1A1 poor metabolizers, and 600 mg/day and 804 ng/mL, respectively, in the other patients. nilotinib 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 24295085-7 2014 Substrates, inhibitors or inducers of UGT1A1 and CYP3A may also affect the pharmacokinetic profile of indacaterol. indacaterol 102-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 24256624-7 2014 Propofol (UGT1A9 inhibitor) effectively blocked G4 generation in HLMs (IC50 63.7 +- 11.6 microM), whereas the UGT1A1 inhibitor bilirubin only produced partial inhibition in HLMs and HIMs. Bilirubin 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 24096683-1 2014 PURPOSE: Dolutegravir (DTG), an unboosted HIV integrase inhibitor (INI), is metabolized by UGT1A1 and to a minor extent by CYP3A. dolutegravir 9-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 24096683-1 2014 PURPOSE: Dolutegravir (DTG), an unboosted HIV integrase inhibitor (INI), is metabolized by UGT1A1 and to a minor extent by CYP3A. dolutegravir 23-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 24642571-0 2014 FOLFIRI combined with bevacizumab as first-line treatment for metastatic colorectal cancer patients with hyperbilirubinemia after UGT1A1 genotyping. FOLFIRI regimen 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 24642571-1 2014 OBJECTIVE: To report a metastatic colorectal cancer patient with hyperbilirubinemia treated with a combination of bevacizumab and FOLFIRI (5-fluorouracil, leucovorin, and irinotecan) using uridine diphosphate glucuronosyl transferase (UGT1A1) genotyping. FOLFIRI regimen 130-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 235-241 24195503-11 2014 Analysis of UGT1A1 alleles may be the basis for modified dosing and reducing the potential toxicity of irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 24523126-13 2014 Examples of adaptive nontoxic changes in liver function, which may elevate direct (conjugated) and/or indirect (unconjugated) bilirubin above baseline levels, include reversible inhibition of UGT1A1-mediated bilirubin metabolism and OATP1B1-, OATP1B3-, or MRP2-mediated transport (Keogh. Bilirubin 126-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 24523126-13 2014 Examples of adaptive nontoxic changes in liver function, which may elevate direct (conjugated) and/or indirect (unconjugated) bilirubin above baseline levels, include reversible inhibition of UGT1A1-mediated bilirubin metabolism and OATP1B1-, OATP1B3-, or MRP2-mediated transport (Keogh. Bilirubin 208-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-104 24195516-2 2014 The metabolism of CPT-11 is mainly controlled by carboxy-lesterase (CES), UDP-glucuronosyltransferase 1A (UGT1A), and beta-glucuronidase (GUSB). Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-111 24329186-0 2014 Evaluation of the effect of UGT1A1 polymorphisms on dolutegravir pharmacokinetics. dolutegravir 52-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 24195516-11 2014 The promoter of the UGT1A1 gene in colorectal cancer cells is methylated, which is an important mechanism of UGT1A1 gene silencing and can be regarded as the target point of research for CPT-11 drug resistance and control mechanisms for the reversal of drug resistance. Irinotecan 187-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 24329186-1 2014 AIM: To evaluate potential pharmacogenetic effects of UGT1A1 polymorphisms on the pharmacokinetics (PK) of dolutegravir (Tivicay ; ViiV Healthcare, NC, USA), an HIV-1 integrase inhibitor. dolutegravir 107-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 24601226-6 2014 At 100 microM of bakuchiol, the activity of UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9 and 1A10 was inhibited by -46.2%, 74.7%, 17.8%, 98.7%, 70.4%, 99.2%, 75.8%, and 93.3%, respectively. bakuchiol 17-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 24329186-4 2014 CONCLUSION: Increased dolutegravir exposure in carriers of UGT1A1 reduced function polymorphisms is not clinically significant based on accumulated safety data so dose adjustment in these individuals is not required. dolutegravir 22-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 24063841-1 2013 Whether conjugated bilirubin concentration, resulting from hepatic UDP-glucuronosyltransferase 1 A1 activity, is associated with cardiovascular disease is unknown. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-99 25147562-4 2014 Human hepatocytes (HH) and HepG2 cells sustained similar treatments, except that the expression of the bilirubin conjugating UDP-glucuronosyltransferase (UGT) 1A1 enzyme and multidrug resistance-associated protein (MRP) 2 transporter was analyzed. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-162 24217933-0 2014 Role of UGT1A1 and ADH gene polymorphisms in pegvisomant-induced liver toxicity in acromegalic patients. pegvisomant 45-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 24088669-0 2013 Polymorphisms in the UGT1A1 gene predict adverse effects of irinotecan in the treatment of gynecologic cancer in Japanese patients. Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 24349450-7 2013 We also found that BPAF glucuronidation could be mediated through several human recombinant UDP-glucuronosyltransferases (UGTs) including UGT1A1, UGT1A3, UGT1A8, UGT1A9, UGT2B4, UGT2B7, UGT2B15 and UGT2B17, among which UGT2B7 showed the highest efficiency of glucuronidation. 4,4'-hexafluorisopropylidene diphenol 19-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 24033692-0 2013 UGT1A1*6, 1A7*3, and 1A9*22 genotypes predict severe neutropenia in FOLFIRI-treated metastatic colorectal cancer in two prospective studies in Japan. FOLFIRI regimen 68-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24033692-1 2013 UNLABELLED: Retrospective studies have suggested that UDP-glucuronosyltransferase (UGT)1A1, UGT1A7, and UGT1A9 predict severe toxicity and efficacy of irinotecan-containing regimens. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-90 24114122-0 2013 UGT1A1 genotype-guided phase I study of irinotecan, oxaliplatin, and capecitabine. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24114122-0 2013 UGT1A1 genotype-guided phase I study of irinotecan, oxaliplatin, and capecitabine. Oxaliplatin 52-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24114122-0 2013 UGT1A1 genotype-guided phase I study of irinotecan, oxaliplatin, and capecitabine. Capecitabine 69-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24114122-11 2013 UGT1A1 *1/*28 and *1/*1 patients treated with IRIN (150) had similar AUCs for the active irinotecan metabolite, SN38 (366 +/- 278 and 350 +/- 159 ng/ml*hr, respectively). Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24114122-14 2013 CONCLUSIONS: UGT1A1 genotype affects the dose and pharmacokinetics of the CAPIRINOX regimen and UGT1A1 genotype-guided dosing of CAPIRINOX is ongoing in a phase II study of small bowel cancer (NCT00433550). capirinox 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24114122-14 2013 CONCLUSIONS: UGT1A1 genotype affects the dose and pharmacokinetics of the CAPIRINOX regimen and UGT1A1 genotype-guided dosing of CAPIRINOX is ongoing in a phase II study of small bowel cancer (NCT00433550). capirinox 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 24088669-6 2013 Homozygosity for UGT1A1*6/*6 and heterozygosity for UGT1A1*6/*28 were associated with an increased risk of absolute neutrophil count and/or diarrhea in Japanese gynecologic cancer patients, despite the lower doses of irinotecan used in these patients. Irinotecan 217-227 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 24088669-7 2013 UGT1A1*6 and UGT1A1*28 are potential predictors of severe absolute neutrophil decrease and diarrhea caused by low-dose irinotecan in gynecologic cancer patients. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24088669-1 2013 Irinotecan is a key chemotherapeutic drug used to treat many tumors, including cervical and ovarian cancers; however, irinotecan can cause toxicity, particularly in the presence of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphisms, which are associated with reduced enzyme activity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 24088669-7 2013 UGT1A1*6 and UGT1A1*28 are potential predictors of severe absolute neutrophil decrease and diarrhea caused by low-dose irinotecan in gynecologic cancer patients. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 24088669-1 2013 Irinotecan is a key chemotherapeutic drug used to treat many tumors, including cervical and ovarian cancers; however, irinotecan can cause toxicity, particularly in the presence of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene polymorphisms, which are associated with reduced enzyme activity. Irinotecan 118-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 24088669-2 2013 Here, we investigated the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28) and their relationships with irinotecan-induced adverse events in patients with gynecologic cancer, who are treated with lower doses of irinotecan than patients with other types of solid tumors. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 24088669-2 2013 Here, we investigated the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28) and their relationships with irinotecan-induced adverse events in patients with gynecologic cancer, who are treated with lower doses of irinotecan than patients with other types of solid tumors. Irinotecan 247-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 24057187-1 2013 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is an important metabolic process during which steroids are converted to more easily excreted compounds in steroid target tissues, such as the prostate. Steroids 129-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-64 24057187-1 2013 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is an important metabolic process during which steroids are converted to more easily excreted compounds in steroid target tissues, such as the prostate. Steroids 129-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 24057187-1 2013 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is an important metabolic process during which steroids are converted to more easily excreted compounds in steroid target tissues, such as the prostate. Steroids 129-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-64 24057187-1 2013 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is an important metabolic process during which steroids are converted to more easily excreted compounds in steroid target tissues, such as the prostate. Steroids 129-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 24107802-0 2013 Hyperbilirubinemia in pazopanib- or sunitinib-treated patients in COMPARZ is associated with UGT1A1 polymorphisms. pazopanib 22-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 24244442-0 2013 UDP-glucuronosyltransferase 1A compromises intracellular accumulation and anti-cancer effect of tanshinone IIA in human colon cancer cells. tanshinone 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 24129235-8 2013 The SU11274 significantly decreased the expression of UGT1A1, but not ABCG2 and ABCB1. ((3Z)-N-(3-chlorophenyl)-3-((3,5-dimethyl-4-((4-methylpiperazin-1-yl)carbonyl)-1H-pyrrol-2-yl)methylene)-N-methyl-2-oxo-2,3-dihydro-1H-indole-5-sulfonamide) 4-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 24107802-0 2013 Hyperbilirubinemia in pazopanib- or sunitinib-treated patients in COMPARZ is associated with UGT1A1 polymorphisms. Sunitinib 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 23950218-1 2013 UDP-glucuronosyltransferase (UGT) 1A1 is the sole enzyme that can metabolize bilirubin. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23950218-7 2013 When HaCaT cells were treated with UVB-exposed tryptophan, UGT1A1 mRNA and activity were significantly induced. Tryptophan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 23950218-8 2013 Treatment of the HaCaT cells with 6-formylindolo[3,2-b]carbazole, which is one of the tryptophan derivatives formed by UVB, resulted in an induction of UGT1A1 mRNA and activity. 6-formylindolo(3,2-b)carbazole 34-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 23950218-8 2013 Treatment of the HaCaT cells with 6-formylindolo[3,2-b]carbazole, which is one of the tryptophan derivatives formed by UVB, resulted in an induction of UGT1A1 mRNA and activity. Tryptophan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 24204915-1 2013 Present study was aimed to explore the effect of (TA)n UGT1A1 gene promoter polymorphism on bilirubin metabolism, bilirubinaemia, predisposition to cholelithiasis and subsequent cholecystectomy, in Sickle-Cell Anemia (SCA) and beta-Thalasemia major (bTH) in Kuwaiti subjects compared to other population. Bilirubin 92-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 24104695-3 2013 Oleic acid, linoleic acid, and docosahexaenoic acid (DHA) strongly inhibited UGT1A1 activity. Docosahexaenoic Acids 31-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24047243-8 2013 UGT1A1 catalyzed the glucuronidation of B[a]P-7,8-catechol and generated two isomeric O-monoglucuronsyl-B[a]P-7,8-catechol products that were identified by RP-HPLC and by LC-MS/MS. 7,8-catechol 46-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24047243-8 2013 UGT1A1 catalyzed the glucuronidation of B[a]P-7,8-catechol and generated two isomeric O-monoglucuronsyl-B[a]P-7,8-catechol products that were identified by RP-HPLC and by LC-MS/MS. o-monoglucuronsyl-b 86-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24047243-8 2013 UGT1A1 catalyzed the glucuronidation of B[a]P-7,8-catechol and generated two isomeric O-monoglucuronsyl-B[a]P-7,8-catechol products that were identified by RP-HPLC and by LC-MS/MS. 7,8-catechol 110-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24104695-0 2013 Impact of fatty acids on human UDP-glucuronosyltransferase 1A1 activity and its expression in neonatal hyperbilirubinemia. Fatty Acids 10-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-62 24104695-3 2013 Oleic acid, linoleic acid, and docosahexaenoic acid (DHA) strongly inhibited UGT1A1 activity. Docosahexaenoic Acids 53-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24104695-2 2013 Here, the impact of fatty acids on human UDP-glucuronosyltransferase (UGT) 1A1--the sole enzyme that can metabolize bilirubin--were examined. Fatty Acids 20-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-78 24104695-4 2013 Forty-eight hours after a treatment with a lower concentration of DHA (10 mg/kg), total bilirubin significantly increased in neonatal hUGT1 mice, which are human neonatal jaundice models. Docosahexaenoic Acids 66-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-139 24104695-2 2013 Here, the impact of fatty acids on human UDP-glucuronosyltransferase (UGT) 1A1--the sole enzyme that can metabolize bilirubin--were examined. Bilirubin 116-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-78 24104695-3 2013 Oleic acid, linoleic acid, and docosahexaenoic acid (DHA) strongly inhibited UGT1A1 activity. Oleic Acid 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24104695-4 2013 Forty-eight hours after a treatment with a lower concentration of DHA (10 mg/kg), total bilirubin significantly increased in neonatal hUGT1 mice, which are human neonatal jaundice models. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-139 24104695-5 2013 In contrast, treatments with higher concentrations of fatty acids (0.1-10 g/kg) resulted in a decrease in serum bilirubin in hUGT1 mice. Fatty Acids 54-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-130 24104695-3 2013 Oleic acid, linoleic acid, and docosahexaenoic acid (DHA) strongly inhibited UGT1A1 activity. Linoleic Acid 12-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 24104695-5 2013 In contrast, treatments with higher concentrations of fatty acids (0.1-10 g/kg) resulted in a decrease in serum bilirubin in hUGT1 mice. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-130 24104695-6 2013 It was further demonstrated that the treatment with higher concentrations of fatty acids induced UGT1A1, possibly by activation of peroxisome proliferator-activated receptors. Fatty Acids 77-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 24104695-7 2013 Our data indicates that activation of peroxisome proliferator-activated receptors would increase UGT1A1 expression, resulting in reduction of serum bilirubin levels in human infants. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 23782005-11 2013 There was a significant association between UGT1A1 genotype and bilirubin grade in the maternal population (p=0.0006) but not neonates (p=0.49). Bilirubin 64-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 23917319-6 2013 Glucuronide conjugates of rilpivirine and a monomethylhydroxylated metabolite of rilpivirine were also detected and were found to be formed by UDP-glucuronosyltransferases (UGTs) UGT1A4 and UGT1A1, respectively. Glucuronides 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 23917319-6 2013 Glucuronide conjugates of rilpivirine and a monomethylhydroxylated metabolite of rilpivirine were also detected and were found to be formed by UDP-glucuronosyltransferases (UGTs) UGT1A4 and UGT1A1, respectively. Rilpivirine 26-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 23917319-6 2013 Glucuronide conjugates of rilpivirine and a monomethylhydroxylated metabolite of rilpivirine were also detected and were found to be formed by UDP-glucuronosyltransferases (UGTs) UGT1A4 and UGT1A1, respectively. Rilpivirine 81-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 23401472-1 2013 Human uridine 5"-diphospho-glucuronosyltransferase (UGT) 1A1 catalyzes the metabolism of numerous clinically and pharmacologically important compounds, such as bilirubin and SN-38. Bilirubin 160-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-60 23401472-1 2013 Human uridine 5"-diphospho-glucuronosyltransferase (UGT) 1A1 catalyzes the metabolism of numerous clinically and pharmacologically important compounds, such as bilirubin and SN-38. Irinotecan 174-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-60 23401472-4 2013 Bisulfite sequence analysis revealed that the CpG-rich region near the UGT1A1 promoter (-85 to +40) was hypermethylated (83%) in the kidney, whereas it was hypomethylated (37%) in the liver. hydrogen sulfite 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 23401472-7 2013 Treatment with 5-aza-2"-deoxycytidine (5-aza-dC), an inhibitor of DNA methylation, resulted in an increase of UGT1A1 mRNA expression in both cell types, but the increase was much larger in HK-2 cells than in HuH-7 cells. Decitabine 15-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 23401472-7 2013 Treatment with 5-aza-2"-deoxycytidine (5-aza-dC), an inhibitor of DNA methylation, resulted in an increase of UGT1A1 mRNA expression in both cell types, but the increase was much larger in HK-2 cells than in HuH-7 cells. Decitabine 39-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 23401472-8 2013 The transfection of an HNF1alpha expression plasmid into the HK-2 cells resulted in an increase of UGT1A1 mRNA only in the presence of 5-aza-dC. Decitabine 135-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 24522354-1 2013 BACKGROUND: In Europeans the TATA box TA7 repeat promoter variant in the UGT1A1 gene (UGT1A1*28) is the major determinant of bilirubin levels. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 23932761-3 2013 Nonetheless, recent Mendelian randomization analyses reveal that individuals who carry low expression alleles of the hepatic bilirubin conjugating enzyme UGT1A1, and hence have somewhat elevated levels of plasma bilirubin throughout life, are not at decreased risk for vascular disorders. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 23932761-3 2013 Nonetheless, recent Mendelian randomization analyses reveal that individuals who carry low expression alleles of the hepatic bilirubin conjugating enzyme UGT1A1, and hence have somewhat elevated levels of plasma bilirubin throughout life, are not at decreased risk for vascular disorders. Bilirubin 212-221 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 24522354-1 2013 BACKGROUND: In Europeans the TATA box TA7 repeat promoter variant in the UGT1A1 gene (UGT1A1*28) is the major determinant of bilirubin levels. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 24065680-2 2013 These mutations result in the deficiency of UGT1A1, a hepatic enzyme essential for bilirubin conjugation. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 24065680-4 2013 Molecular genetic analysis showed a homozygous UGT1A1 promoter mutation [A(TA)7TAA] and a heterozygous insertion of 1 adenosine nucleotide between positions 353 and 354 in exon 1 of UGT1A1 that caused a frameshift with a premature stop codon. adenosine nucleotide 118-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 23750830-16 2013 These data suggest that the proposed surrogate probe substrates to evaluate the in vitro inhibition of UGT1A1, OATP1B1, and BSEP may be suitable to assess bilirubin disposition. Bilirubin 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 23726867-6 2013 Up-regulation of metabolic enzyme genes provided evidence for the involvement of phase I (CYP1A1, CYP1B1, ALDH1A2 and CES2) and phase II (UGT1A6, UGT1A1, NAT1 and GSTM3) enzymes in the detoxification response and potential activation of CYN. cylindrospermopsin 237-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 23677771-10 2013 Axitinib is metabolized primarily in the liver by cytochrome P450 (CYP) 3A4/5 and, to a lesser extent (<10 % each), by CYP1A2, CYP2C19, and uridine diphosphate glucuronosyltransferase (UGT) 1A1. Axitinib 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-196 23892411-0 2013 Analysis of UGT1A1*28 genotype and SN-38 pharmacokinetics for irinotecan-based chemotherapy in patients with advanced colorectal cancer: results from a multicenter, retrospective study in Shanghai. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23892411-1 2013 BACKGROUND: The UGT1A1*28 polymorphism, although closely linked with CPT-11-related adverse effects, cannot be used alone to guide individualized treatment decisions. Irinotecan 69-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 23892411-6 2013 To understand differences in genotype expression, the frequency of UGT1A1*28 thymine-adenine (TA) repeats in TATA box arrangement was assessed by PCR with genomic DNA extracted from peripheral blood. thymine-adenine 77-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 23892411-6 2013 To understand differences in genotype expression, the frequency of UGT1A1*28 thymine-adenine (TA) repeats in TATA box arrangement was assessed by PCR with genomic DNA extracted from peripheral blood. Tantalum 94-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 24147345-2 2013 in vitro incubation method using recombinant UGTs-catalyzed 4-methylumbelliferone (4-MU) glucuronidation was used to evaluate the inhibition towards important UGT isoforms in the liver, including UGT1A1, 1A3, 1A6, 1A9, and 2B7. Hymecromone 83-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 23786524-0 2013 Mutual regioselective inhibition of human UGT1A1-mediated glucuronidation of four flavonoids. Flavonoids 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 23669385-8 2013 An inactive ether glucuronide, formed primarily via UGT1A1, was the principal biotransformation product at 18.9% of the dose excreted in urine and the principal metabolite in plasma. ether glucuronide 12-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 23786524-1 2013 UDP-glucuronosyltransferase (UGT) 1A1-catalyzed glucuronidation is an important elimination pathway of flavonoids, and mutually inhibitory interactions may occur when two or more flavonoids are coadministered. Flavonoids 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23786524-1 2013 UDP-glucuronosyltransferase (UGT) 1A1-catalyzed glucuronidation is an important elimination pathway of flavonoids, and mutually inhibitory interactions may occur when two or more flavonoids are coadministered. Flavonoids 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23786524-2 2013 Our recent research suggested that glucuronidation of flavonoids displayed distinct positional preferences, but whether this will lead to the mutually regioselective inhibition of UGT1A1-mediated glucuronidation of flavonoids is unknown. Flavonoids 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 23786524-2 2013 Our recent research suggested that glucuronidation of flavonoids displayed distinct positional preferences, but whether this will lead to the mutually regioselective inhibition of UGT1A1-mediated glucuronidation of flavonoids is unknown. Flavonoids 215-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 23689708-1 2013 Atazanavir (ATV) causes an elevation of unconjugated hyperbilirubinemia (HBR) as a result of UDP glucuronyltransferase (UGT) 1A1 inhibition. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-128 23898114-0 2013 UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism-directed phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23898114-0 2013 UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism-directed phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer. doxifluridine 97-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23689708-1 2013 Atazanavir (ATV) causes an elevation of unconjugated hyperbilirubinemia (HBR) as a result of UDP glucuronyltransferase (UGT) 1A1 inhibition. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-128 23898114-0 2013 UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism-directed phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer. doxifluridine 123-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23898114-1 2013 AIM: We performed a phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer based on UDP-glucuronosyltransferase (UGT) 1A1 polymorphism. Irinotecan 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-168 23800356-0 2013 Recurrent cervical cancer in a patient who was compound heterozygous for UGT1A1*6 and UGT1A1*28 presenting with serious adverse events during irinotecan hydrochloride/nedaplatin therapy. Irinotecan 142-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 23898114-1 2013 AIM: We performed a phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer based on UDP-glucuronosyltransferase (UGT) 1A1 polymorphism. doxifluridine 54-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-168 23898114-1 2013 AIM: We performed a phase II study of irinotecan with 5"-deoxy-5-fluorouridine (5"-DFUR) for metastatic colorectal cancer based on UDP-glucuronosyltransferase (UGT) 1A1 polymorphism. doxifluridine 80-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-168 23800356-0 2013 Recurrent cervical cancer in a patient who was compound heterozygous for UGT1A1*6 and UGT1A1*28 presenting with serious adverse events during irinotecan hydrochloride/nedaplatin therapy. Irinotecan 142-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 224-230 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. Irinotecan 162-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 23800356-6 2013 The findings in this patient indicate that CPT-11 should be administered with great care, even at a dose of 60 mg/m2, in patients receiving combined therapy with CPT-11 and NDP who are compound heterozygous for UGT1A1*6 and UGT1A1*28. nedaplatin 173-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 211-217 23926009-1 2013 OBJECTIVE: To analyze potential mutations of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene in patients with unconjugated hyperbilirubinemia, and to explore the correlation between the mutations and total serum bilirubin levels. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-92 23580084-0 2013 UGT1A1*28 is associated with decreased systemic exposure of atorvastatin lactone. Atorvastatin lactone 60-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23580084-2 2013 Atorvastatin acid is converted to its corresponding lactone form spontaneously or via glucuronidation mediated by uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A3. Atorvastatin 0-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-175 23580084-2 2013 Atorvastatin acid is converted to its corresponding lactone form spontaneously or via glucuronidation mediated by uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A3. Lactones 52-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-175 23580084-8 2013 RESULTS: Carriers of the low-expression allele UGT1A1*28(TA) 7 tended to have lower levels of atorvastatin lactone (p < 0.05) than carriers with the normal-activity allele (TA) 6 . Atorvastatin lactone 94-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 23580084-9 2013 CONCLUSION: The low-expression UGT1A1*28(TA) 7 allele seems to be associated with decreased systemic exposure of the suspected muscle-toxic metabolite atorvastatin lactone. Atorvastatin lactone 151-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 23065713-4 2013 Data fitting using Dixon and Lineweaver-Burk plots demonstrated the competitive inhibition of liquiritigenin towards UGT1A1 and UGT1A9-mediated 4-MU glucuronidation reaction. liquiritigenin 94-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 23926009-1 2013 OBJECTIVE: To analyze potential mutations of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene in patients with unconjugated hyperbilirubinemia, and to explore the correlation between the mutations and total serum bilirubin levels. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 23926009-8 2013 CONCLUSION: The level of total serum bilirubin is correlated with the number of UGT1A1 gene mutations as well as their heterozygous or homozygous status. Bilirubin 37-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 23606168-5 2013 BCL11A and XmnI polymorphisms had significant effects on baseline HbF, while UGT1A1 promoter polymorphisms significantly influenced baseline serum bilirubin. Bilirubin 147-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 23753274-4 2013 Second, taking advantage of mendelian randomization, we tested whether a genetic variant in the bilirubin glucoronidating enzyme UGT1A1 (rs6742078) was associated with increased plasma bilirubin levels and, in turn, with an increased risk of symptomatic gallstone disease. Bilirubin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 23753274-4 2013 Second, taking advantage of mendelian randomization, we tested whether a genetic variant in the bilirubin glucoronidating enzyme UGT1A1 (rs6742078) was associated with increased plasma bilirubin levels and, in turn, with an increased risk of symptomatic gallstone disease. Bilirubin 185-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 23753274-8 2013 UGT1A1 genotype explained 20% of the total variation in plasma bilirubin levels and was associated with increases in the mean plasma bilirubin level overall of +16% (+0.09 mg/dL) in GT heterozygotes and +90% (+0.50 mg/dL) in TT homozygotes compared with GG homozygotes, with similar effects in women and men (P for trend <.001 for all). Bilirubin 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23753274-8 2013 UGT1A1 genotype explained 20% of the total variation in plasma bilirubin levels and was associated with increases in the mean plasma bilirubin level overall of +16% (+0.09 mg/dL) in GT heterozygotes and +90% (+0.50 mg/dL) in TT homozygotes compared with GG homozygotes, with similar effects in women and men (P for trend <.001 for all). Bilirubin 133-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23670235-5 2013 RESULTS: UGT1A1, UGT1A4, UGT1A8, UGT2B7, and SULT1A1 were found to be involved in the formation of inactive ABT-751 glucuronide (ABT-751G) and sulfate (ABT-751S). 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 108-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 24060717-0 2013 UGT1A1 promoter polymorphism associated with serum bilirubin level in Saudi patients with sickle cell disease. Bilirubin 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 24060717-1 2013 BACKGROUND AND OBJECTIVES: Polymorphism in (TA)n of the UGT1A1 promoter influences bilirubin level and risk of gallstones in patients with sickle cell disease (SCD) of African descent. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 24060717-8 2013 Increased (TA)n of the UGT1A1 promoter (P < .0001), male gender (P=.02), higher LDH (P=.001), and lower Hb level (P=.009) were associated with higher bilirubin level, while the co-inheritance of a-thalassemia (P=.003) was linked with lower bilirubin level. Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 24060717-8 2013 Increased (TA)n of the UGT1A1 promoter (P < .0001), male gender (P=.02), higher LDH (P=.001), and lower Hb level (P=.009) were associated with higher bilirubin level, while the co-inheritance of a-thalassemia (P=.003) was linked with lower bilirubin level. Bilirubin 243-252 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 24060717-12 2013 CONCLUSION: (TA)n in the UGT1A1 promoter and intensity of hemolysis modify steady-state serum bilirubin level in SCD. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 23682072-4 2013 Using homogenates from HEK293 UGT-overexpressing cell lines, raloxifene was shown to be glucuronidated primarily by the hepatic UGTs 1A1 and 1A9 and the extra-hepatic UGTs 1A8 and 1A10; no detectable raloxifene glucuronidation activity was found for UGT2B enzymes. Raloxifene Hydrochloride 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-144 23682072-5 2013 Functional UGT1A1 transcriptional promoter genotypes were significantly (Ptrend = 0.005) associated with ral-6-Gluc formation in human liver microsomes, and, consistent with the decreased raloxifene glucuronidation activities observed in vitro with cell lines overexpressing UGT1A8 variants, the UGT1A8*2 variant was significantly (P = 0.023) correlated with total raloxifene glucuronide formation in human jejunum homogenates. Raloxifene Hydrochloride 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 23682072-5 2013 Functional UGT1A1 transcriptional promoter genotypes were significantly (Ptrend = 0.005) associated with ral-6-Gluc formation in human liver microsomes, and, consistent with the decreased raloxifene glucuronidation activities observed in vitro with cell lines overexpressing UGT1A8 variants, the UGT1A8*2 variant was significantly (P = 0.023) correlated with total raloxifene glucuronide formation in human jejunum homogenates. Raloxifene Hydrochloride 365-375 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 23682072-5 2013 Functional UGT1A1 transcriptional promoter genotypes were significantly (Ptrend = 0.005) associated with ral-6-Gluc formation in human liver microsomes, and, consistent with the decreased raloxifene glucuronidation activities observed in vitro with cell lines overexpressing UGT1A8 variants, the UGT1A8*2 variant was significantly (P = 0.023) correlated with total raloxifene glucuronide formation in human jejunum homogenates. Glucuronides 376-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 23670235-5 2013 RESULTS: UGT1A1, UGT1A4, UGT1A8, UGT2B7, and SULT1A1 were found to be involved in the formation of inactive ABT-751 glucuronide (ABT-751G) and sulfate (ABT-751S). Glucuronides 116-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23670235-5 2013 RESULTS: UGT1A1, UGT1A4, UGT1A8, UGT2B7, and SULT1A1 were found to be involved in the formation of inactive ABT-751 glucuronide (ABT-751G) and sulfate (ABT-751S). 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23670235-5 2013 RESULTS: UGT1A1, UGT1A4, UGT1A8, UGT2B7, and SULT1A1 were found to be involved in the formation of inactive ABT-751 glucuronide (ABT-751G) and sulfate (ABT-751S). Sulfates 143-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23670235-5 2013 RESULTS: UGT1A1, UGT1A4, UGT1A8, UGT2B7, and SULT1A1 were found to be involved in the formation of inactive ABT-751 glucuronide (ABT-751G) and sulfate (ABT-751S). 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23595344-10 2013 CONCLUSION: Irinotecan dosing based on UGT1A1*28 and *6 is feasible, and higher doses of irinotecan can be safely administered in patients with 0 or 1 DA, compared to those with 2 DA. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23713759-6 2013 Moreover, the A-ring OH-4 group promoted glucuronidation at the 2" position for the reaction of 4,2"-dihydroxychalcone with UGT1A1 and 1A3. 4,2'-DIHYDROXYCHALCONE 96-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-138 23840132-0 2013 Severe irinotecan-induced toxicity in a patient with UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 23840132-0 2013 Severe irinotecan-induced toxicity in a patient with UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 23840132-1 2013 Many studies have demonstrated the impact of UGT1A1 on toxicity of irinotecan. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 23840132-3 2013 Based on this, prescribers of irinotecan are advised that patients with UGT1A1 28 (TA 7/7) should start with a reduced dose of irinotecan, although a particular dose is not specified. Irinotecan 30-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 23840132-3 2013 Based on this, prescribers of irinotecan are advised that patients with UGT1A1 28 (TA 7/7) should start with a reduced dose of irinotecan, although a particular dose is not specified. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 23840132-4 2013 Research in Asian countries has shown a lower incidence of UGT1A1 28 (TA 7/7), while UGT1A1 6 (A/A) is more often found and is associated with severe irinotecan-related neutropenia. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23840132-7 2013 Simultaneous heterozygous UGT1A1 28 and UGT1A1 6 polymorphisms may produce higher exposure to SN-38 and a higher risk of adverse effects related to irinotecan. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 23840132-8 2013 Additional studies will be necessary to determine the optimal starting dose of irinotecan for patients with both UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 23840132-8 2013 Additional studies will be necessary to determine the optimal starting dose of irinotecan for patients with both UGT1A1 28 and UGT1A1 6 polymorphisms. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 23595344-0 2013 A UGT1A1*28 and *6 genotype-directed phase I dose-escalation trial of irinotecan with fixed-dose capecitabine in Korean patients with metastatic colorectal cancer. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 23595344-0 2013 A UGT1A1*28 and *6 genotype-directed phase I dose-escalation trial of irinotecan with fixed-dose capecitabine in Korean patients with metastatic colorectal cancer. Capecitabine 97-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 23595344-1 2013 PURPOSE: UGT1A1 genotypes are important when considering treatment with irinotecan-containing regimens. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 23595344-2 2013 In this study, we determined the dose, efficacy, and tolerability of irinotecan according to UGT1A1 genotypes when combined with capecitabine in patients with metastatic colorectal cancer. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 23559402-9 2013 Polymorphisms in CYP2C9, TPMT, UGT1A1, MDR1, and 5-HTR2A were related to some adverse effects of olanzapine. Olanzapine 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 23241680-0 2013 Microarray with LNA-probes for genotyping of polymorphic variants of Gilbert"s syndrome gene UGT1A1(TA)n. BACKGROUND: Gilbert"s syndrome is a common metabolic dysfunction characterized by elevated levels of unconjugated bilirubin in the bloodstream. Bilirubin 220-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 23537215-0 2013 The association between heterozygosity for UGT1A1*6, UGT1A1*28, and variation in the serum total-bilirubin level in healthy young Japanese adults. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 23537215-10 2013 Multiple regression analysis showed significant relationships between the serum total-bilirubin level, the UGT1A1 genotypes *1/*28 and *6/*28, and sex. Bilirubin 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 23537215-12 2013 CONCLUSIONS: We found that the UGT1A1 genotypes *1/*28 and *6/*28 as well as sex contributed to interindividual variations in the serum total-bilirubin levels. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 23537215-13 2013 In contrast, UGT1A1*1/*6 showed only minimal involvement in individual differences in serum total-bilirubin levels. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 23647681-1 2013 OBJECTIVES: Interindividual variability in glucuronidation of bilirubin and drugs by UDP-glucuronosyltransferase 1A1 (UGT1A1) is considerable and only partially explained by genetic polymorphisms and enzyme inducers. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-116 23647681-1 2013 OBJECTIVES: Interindividual variability in glucuronidation of bilirubin and drugs by UDP-glucuronosyltransferase 1A1 (UGT1A1) is considerable and only partially explained by genetic polymorphisms and enzyme inducers. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 23647681-6 2013 Multivariate regression analysis identified *28/*28 genotype, -4 CpG site methylation and alcohol history as significant predictors of UGT1A1 protein content. Alcohols 90-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23647681-7 2013 Exclusion of livers with *28/*28 genotype or alcohol history revealed positive correlations of -4 CpG methylation with bilirubin glucuronidation (R = 0.73, P < 0.00001) and UGT1A1 protein content (R = 0.54, P = 0.008). Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 23737969-10 2013 Body weight >=40 kg and homozygosity for UGT1A1*6 were risk factors of creatinine elevation (OR = 8.48, 95% CI = 1.7-43.57, P = 0.010 and OR = 14.17, 95% CI = 1.34-150.35, P = 0.028). Creatinine 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 23387445-0 2013 Phase I and II metabolism and MRP2-mediated export of bosentan in a MDCKII-OATP1B1-CYP3A4-UGT1A1-MRP2 quadruple-transfected cell line. Bosentan 54-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 23387445-8 2013 Additionally, a direct glucuronide of bosentan could be identified intracellularly in cell lines expressing UGT1A1 and in the apical compartments of cell lines expressing UGT1A1 and MRP2. Glucuronides 23-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 23387445-8 2013 Additionally, a direct glucuronide of bosentan could be identified intracellularly in cell lines expressing UGT1A1 and in the apical compartments of cell lines expressing UGT1A1 and MRP2. Glucuronides 23-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 23387445-8 2013 Additionally, a direct glucuronide of bosentan could be identified intracellularly in cell lines expressing UGT1A1 and in the apical compartments of cell lines expressing UGT1A1 and MRP2. Bosentan 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 23387445-8 2013 Additionally, a direct glucuronide of bosentan could be identified intracellularly in cell lines expressing UGT1A1 and in the apical compartments of cell lines expressing UGT1A1 and MRP2. Bosentan 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 23387445-9 2013 CONCLUSIONS AND IMPLICATIONS: These in vitro data indicate that bosentan is a substrate of UGT1A1. Bosentan 64-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 23090703-2 2013 This study was performed to examine whether common polymorphisms in UGT1A1, UGT2B7, CYP2C19 and ABCB1 affect the PK of labetalol. Labetalol 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 23408116-7 2013 Cotransfection studies demonstrated an inhibitory effect of exon 5b containing cDNAs on acetaminophen glucuronidation by UGT1A1 and UGT1A6 cDNAs containing exon 5a. exon 5b 60-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 23408116-7 2013 Cotransfection studies demonstrated an inhibitory effect of exon 5b containing cDNAs on acetaminophen glucuronidation by UGT1A1 and UGT1A6 cDNAs containing exon 5a. Acetaminophen 88-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 23299247-6 2013 Demethylation was inhibited significantly by furafylline and predominantly catalysed by recombinant CYP1A2, whereas glucuronidation was inhibited by silibinin, quercetin, as well as 1-naphthol and catalysed by recombinant UGT1A1, UGT1A3, UGT1A7, UGT1A8, UGT1A9 and UGT1A10. 1-naphthol 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 23888707-3 2013 Additionally, effects of PA and PC on UGT1A1 mRNA and protein expressions were also measured after transient transfection of a specific CAR siRNA for 72 h in HepG2 cells. praeruptorin C 32-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 23888707-5 2013 The results suggest that PA and PC can significantly up-regulate UGT1A1 expression partially via the CAR-mediated pathway. praeruptorin A 25-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 23888707-5 2013 The results suggest that PA and PC can significantly up-regulate UGT1A1 expression partially via the CAR-mediated pathway. praeruptorin C 32-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 23619273-3 2013 We analyzed the polymorphism A(TA)<formula>_{n}</formula> TAA at the UGT1A1 promoter and the relationships between the various A(TA)<formula>_{n}</formula> TAA genotypes and alleles and bilirubin levels and occurrence of cholelithiasis. Bilirubin 210-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 23548653-0 2013 Effect of the UGT1A1*28 allele on unconjugated hyperbilirubinemia in HIV-positive patients receiving Atazanavir: a systematic review. Atazanavir Sulfate 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 23548653-1 2013 OBJECTIVE: To systematically examine the literature assessing the effect of uridine 5"-diphospho-glucuronosyltransferase (UGT)1A1*28 genetic polymorphisms on atazanavir-associated hyperbilirubinemia. Atazanavir Sulfate 158-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 23090703-10 2013 Future larger studies are needed to evaluate the effect of CYP2C19 and UGT1A1 polymorphisms on the PK of labetalol stereoisomers and the pharmacodynamic effects. Labetalol 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 23230132-8 2013 All recombinant UGTs, except UGT1A1, 1A6, and 1A10, produced EtG in detectable amounts. ethyl glucuronide 61-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 23237922-3 2013 Of these, 33 patients were subjected to HLA typing and polymorphism analyses of CYP3A5, ABCB1, ABCC2, and UGT1A1, which are involved in the metabolism and membrane transport of sorafenib. Sorafenib 177-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 23488780-7 2013 Assessment of a panel of recombinant UGT isoforms revealed that UGT1A1, UGT1A3 and UGT1A9 could catalyse the glucuronidation of arbidol. umifenovir 128-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-0 2013 [Influence of genetic polymorphisms in UGT1A1, UGT1A7 and UGT1A9 on the pharmacokynetics of irinotecan, SN-38 and SN-38G]. Irinotecan 114-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 23789755-1 2013 OBJECTIVE: To evaluate the influence of genetic polymorphism in UGT1A1, UGT1A7 and UGT1A9 on the population pharmacokinetics of irinotecan and its metabolites, SN-38 and SN-38G. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 23789755-7 2013 The presence of UGT1A1*28, UGT1A7*3, UGT1A9*22 genotypes decreases SN-38 clearance between 20 and 36%. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 23789755-9 2013 CONCLUSION: The inclusion of pharmacokinetic-pharmacogenomic information can add value to the individualized dose adjustment of irinotecan, because it will let quantitatively handle dose reductions in patients with iatrogenic toxicity due to UGT1A1 genetic polymorphisms. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 242-248 22813462-4 2013 Recombinant UGT isoforms screening experiment and enzyme kinetic study showed that UGT1A1 completely contributed to the glucuronidation of thienorphine. thienorphine 139-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 23663345-0 2013 [Effects of autophagy modulator on autophagy and uridine 5"-diphospho-glucuronosyltransferase 1A1 induced by sulforaphane]. sulforaphane 109-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-97 23663345-1 2013 OBJECTIVE: To explore the effects of 3-methyladenine (3-MA) and rapamycin (Rapa) on autophagy and uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) induced by sulforaphane (SFN) in human colon cancer Caco-2 cells. 3-methyladenine 54-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-146 23663345-1 2013 OBJECTIVE: To explore the effects of 3-methyladenine (3-MA) and rapamycin (Rapa) on autophagy and uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) induced by sulforaphane (SFN) in human colon cancer Caco-2 cells. sulforaphane 167-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-146 23663345-1 2013 OBJECTIVE: To explore the effects of 3-methyladenine (3-MA) and rapamycin (Rapa) on autophagy and uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1) induced by sulforaphane (SFN) in human colon cancer Caco-2 cells. sulforaphane 181-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-146 23663345-13 2013 And Rapa also potentiates SFN-induced UGT1A1 expression. sulforaphane 26-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 22934695-2 2013 This study aims to investigate the association between UGT1A1 gene polymorphisms and irinotecan related toxicities in Chinese Han gastrointestinal cancer patients receiving FOLFIRI regimen chemotherapy. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 22934695-2 2013 This study aims to investigate the association between UGT1A1 gene polymorphisms and irinotecan related toxicities in Chinese Han gastrointestinal cancer patients receiving FOLFIRI regimen chemotherapy. FOLFIRI regimen 173-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 22934695-12 2013 The baseline serum total bilirubin was elevated in UGT1A1*28, *93 allele carriers and *60 homozygote, but with no relationship with severe toxicities. Bilirubin 25-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 23132334-5 2013 UDP-glucuronosyltransferase (UGT) 1A1 is the main enzyme responsible for the metabolism of DTG in vivo, with cytochrome P450 (P450) 3A4 being a notable pathway and UGT1A3 and UGT1A9 being only minor pathways. dolutegravir 91-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 23148286-0 2013 Impact of UGT1A1 Gilbert variant on discontinuation of ritonavir-boosted atazanavir in AIDS Clinical Trials Group Study A5202. Ritonavir 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 23148286-0 2013 Impact of UGT1A1 Gilbert variant on discontinuation of ritonavir-boosted atazanavir in AIDS Clinical Trials Group Study A5202. Atazanavir Sulfate 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 23148286-1 2013 The UGT1A1*28 variant has been associated with hyperbilirubinemia and atazanavir discontinuation. Atazanavir Sulfate 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 23303296-12 2013 In conclusion, UGT1A1*6/*28, UGT1A7*3/*3, and UGT1A9*1/*1 should be taken into consideration as markers for preventing severe adverse events of CPT-11 administration. Irinotecan 144-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 23277092-4 2013 RESULTS: Two haplotypes in UGT1A1/UGT1A8 were weak predictors of reduced M6G/morphine and M3G/morphine serum ratios after oral administration (false discovery rate-corrected P-values<0.1). Morphine 77-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 23277092-4 2013 RESULTS: Two haplotypes in UGT1A1/UGT1A8 were weak predictors of reduced M6G/morphine and M3G/morphine serum ratios after oral administration (false discovery rate-corrected P-values<0.1). Morphine 94-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 23148825-0 2013 Protein-protein interactions between the bilirubin-conjugating UDP-glucuronosyltransferase UGT1A1 and its shorter isoform 2 regulatory partner derived from alternative splicing. Bilirubin 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 23148825-4 2013 The bilirubin-conjugating UGT1A1 was used as a prototype. Bilirubin 4-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 23388413-2 2013 Among the UGT gene family members, only UGT1A1 is involved in bilirubin conjugation. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Bilirubin 64-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Bilirubin 64-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23130636-2 2013 UGT1A1 (UDP-glucuronosyltransferase 1A1) is a critical gene for bilirubin metabolism and irinotecan detoxification. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-39 23089672-11 2013 Regarding phase II enzyme polymorphism, the anticancer treatment with mercaptopurines and irinotecan is still considered important in relation to the polymorphism of TPMT and UGT1A1, respectively. Mercaptopurine 70-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 175-181 23089672-11 2013 Regarding phase II enzyme polymorphism, the anticancer treatment with mercaptopurines and irinotecan is still considered important in relation to the polymorphism of TPMT and UGT1A1, respectively. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 175-181 24390949-3 2013 Thymine-adenine (TA) repeats in the promoter region of UGT1A1 gene investigated by means of polymerase- chain reaction (PCR) DNA sequencing. thymine-adenine 0-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 23036853-8 2013 Among the enzymes catalyzing BaP oxidation and conjugation, cytochrome P450 1A1, 1A2, 3A4 and UDP-glucuronosyltransferase 1A1 mRNAs were induced by the exposure to TCDD. Benzo(a)pyrene 29-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-125 23036853-8 2013 Among the enzymes catalyzing BaP oxidation and conjugation, cytochrome P450 1A1, 1A2, 3A4 and UDP-glucuronosyltransferase 1A1 mRNAs were induced by the exposure to TCDD. Polychlorinated Dibenzodioxins 164-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-125 23147267-0 2013 UDP-glucuronosyltransferase 1A1 (UGT1A1) gene haplotypes and their effect on serum bilirubin concentration in healthy Indian adults. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 23147267-0 2013 UDP-glucuronosyltransferase 1A1 (UGT1A1) gene haplotypes and their effect on serum bilirubin concentration in healthy Indian adults. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 23147267-1 2013 The aim of the present study was to investigate the allele and genotype frequencies and haplotype structures of the variants in the UGT1A1 gene and their association with serum bilirubin levels in healthy adults. Bilirubin 177-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 23147267-7 2013 Promoter polymorphisms and a common haplotype of the UGT1A1 gene are associated with serum bilirubin concentrations and could be a genetic risk factor for hyperbilirubinemia in Indians. Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 24390949-3 2013 Thymine-adenine (TA) repeats in the promoter region of UGT1A1 gene investigated by means of polymerase- chain reaction (PCR) DNA sequencing. Tantalum 17-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 23089802-3 2013 Impaired or reduced UGT1A1 activity causes unconjugated hyperbilirubinemia (Gilbert"s syndrome and Crigler-Najjar syndrome) and side effects of drug treatment such as SN-38 (active metabolite of the anticancer drug irinotecan)-induced toxicity. Irinotecan 215-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23449330-7 2013 E-3OH and SN-38 glucuronidation by recombinant hUGT1A1 and mpUGT1A1 showed allosteric sigmoidal kinetics. 3oh 2-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-54 23449330-7 2013 E-3OH and SN-38 glucuronidation by recombinant hUGT1A1 and mpUGT1A1 showed allosteric sigmoidal kinetics. Irinotecan 10-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-54 23264445-1 2013 BACKGROUND: Homozygosity for UGT1A1*28/*28 has been reported to be associated with atazanavir-associated hyperbilirubinaemia and premature atazanavir discontinuation. Atazanavir Sulfate 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 23264445-1 2013 BACKGROUND: Homozygosity for UGT1A1*28/*28 has been reported to be associated with atazanavir-associated hyperbilirubinaemia and premature atazanavir discontinuation. Atazanavir Sulfate 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 23264445-4 2013 We assumed the UGT1A1-associated atazanavir discontinuation rate reported in the Swiss HIV Cohort Study (a *28/*28 frequency of 14.9%), equal efficacy and cost of atazanavir and darunavir and a genetic assay cost of $107. Atazanavir Sulfate 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. Estradiol 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. 3oh 56-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. Irinotecan 65-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23449330-5 2013 Then the kinetics of estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation by recombinant UGT1A1s as well as human and minipig liver microsomes were analyzed. Irinotecan 97-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 23053265-0 2013 Uridine diphosphate glucuronosyl transferase 1 family polypeptide A1 gene (UGT1A1) polymorphisms are associated with toxicity and efficacy in irinotecan monotherapy for refractory pancreatic cancer. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 23089802-1 2013 Human UDP-glucuronosyltransferase (UGT) 1A1 is the enzyme that detoxifies neurotoxic bilirubin by conjugating it with glucuronic acid. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 23089802-1 2013 Human UDP-glucuronosyltransferase (UGT) 1A1 is the enzyme that detoxifies neurotoxic bilirubin by conjugating it with glucuronic acid. Glucuronic Acid 118-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 23089802-3 2013 Impaired or reduced UGT1A1 activity causes unconjugated hyperbilirubinemia (Gilbert"s syndrome and Crigler-Najjar syndrome) and side effects of drug treatment such as SN-38 (active metabolite of the anticancer drug irinotecan)-induced toxicity. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23781580-0 2013 UGT1A1 genotype-specific phase I and pharmacokinetic study for combination chemotherapy with irinotecan and cisplatin: a Saitama Tumor Board study. Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23648677-3 2013 A study using UGT-expressing supersomes revealed that sipoglitazar glucuronidation was more extensively catalyzed by UGT1A1, 1A3, 1A6, 2B4, and 2B15 than by other UGTs. sipoglitazar 54-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 23648677-5 2013 In a correlation study between sipoglitazar glucuronidation and UGT isoform-specific activities, the glucuronidation of S-oxazepam, a specific substrate for UGT2B15, strongly correlated with that of sipoglitazar, as compared with that of beta-estradiol, a representative UGT1A1 substrate. sipoglitazar 31-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 23648677-5 2013 In a correlation study between sipoglitazar glucuronidation and UGT isoform-specific activities, the glucuronidation of S-oxazepam, a specific substrate for UGT2B15, strongly correlated with that of sipoglitazar, as compared with that of beta-estradiol, a representative UGT1A1 substrate. Oxazepam 120-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 23648677-5 2013 In a correlation study between sipoglitazar glucuronidation and UGT isoform-specific activities, the glucuronidation of S-oxazepam, a specific substrate for UGT2B15, strongly correlated with that of sipoglitazar, as compared with that of beta-estradiol, a representative UGT1A1 substrate. sipoglitazar 199-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 23781580-0 2013 UGT1A1 genotype-specific phase I and pharmacokinetic study for combination chemotherapy with irinotecan and cisplatin: a Saitama Tumor Board study. Cisplatin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23781580-2 2013 The aim of the present study was to determine the RD of irinotecan in combination with cisplatin (CPT-P) for individuals with or without UGT1A1 polymorphisms. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 23781580-8 2013 CONCLUSION: UGT1A1 genotype-based RDs of irinotecan in CPT-P therapy were determined. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23781580-8 2013 CONCLUSION: UGT1A1 genotype-based RDs of irinotecan in CPT-P therapy were determined. cpt 55-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 23781580-8 2013 CONCLUSION: UGT1A1 genotype-based RDs of irinotecan in CPT-P therapy were determined. Phosphorus 56-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 22805420-7 2013 UGT1A1 rs6742078 TT versus GG genotype was associated with 95% increased bilirubin levels (P < 0.001); TT versus GG genotype was associated with odds ratios (ORs) of 1.03 (95% CI, 0.96-1.11; P = 0.73) for IHD and 1.01 (95% CI, 0.92-1.12; P = 0.68) for MI. Bilirubin 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23237733-2 2013 Therefore, the aim of the present study was to investigate the inhibition of isoliquiritigenin towards important UGT isoforms in the liver and intestine, including UGT1A1, 1A3, 1A6, 1A7, 1A8, 1A9 and 1A10. isoliquiritigenin 77-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 23237733-4 2013 The results showed that 100muM of isoliquiritigenin inhibited the activity of UGT1A1, UGT1A3, UGT1A6, UGT1A7, UGT1A8, UGT1A9, and UGT1A10 by 95.2%, 76.1%, 78.9%, 87.2%, 67.2%, 94.8%, and 91.7%, respectively. isoliquiritigenin 34-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 23783485-0 2013 Associations between UGT1A1*6/*28 polymorphisms and irinotecan-induced severe toxicity in Chinese gastric or esophageal cancer patients. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 23686699-0 2013 UGT1A1 6/28 polymorphisms could predict irinotecan-induced severe neutropenia not diarrhea in Chinese colorectal cancer patients. Irinotecan 40-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23783485-1 2013 The aim of this study was to investigate the associations between UDP-glucuronosyltransferase (UGT) 1A1 polymorphisms and irinotecan-induced toxicities in Chinese advanced gastric or esophageal cancer patients. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-103 23686699-1 2013 The aim of this study was to investigate the associations between UDP-glucuronosyltransferase (UGT) 1A1 polymorphisms and irinotecan-induced toxicities in Chinese advanced colorectal cancer patients. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-103 23686699-9 2013 Both UGT1A1 6 and UGT1A1 28 variants were closely associated with irinotecan-induced severe neutropenia, but not diarrhea. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 23236239-0 2012 UGT1A1 predicts outcome in colorectal cancer treated with irinotecan and fluorouracil. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23686699-9 2013 Both UGT1A1 6 and UGT1A1 28 variants were closely associated with irinotecan-induced severe neutropenia, but not diarrhea. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 23516488-0 2013 Association between UGT1A1*28 polymorphisms and clinical outcomes of irinotecan-based chemotherapies in colorectal cancer: a meta-analysis in Caucasians. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23516488-1 2013 BACKGROUND: Whether UGT1A1*28 genotype is associated with clinical outcomes of irinotecan (IRI)-based chemotherapy in Colorectal cancer (CRC) is an important gap in existing knowledge to inform clinical utility. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 23382909-5 2013 The major pathway of metabolism involves UGT1A1-mediated glucuronidation and a good correlation has been identified between belinostat glucuronide formation and glucuronidation of known UGT1A1 substrates. Belinostat glucuronide 124-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 23382909-5 2013 The major pathway of metabolism involves UGT1A1-mediated glucuronidation and a good correlation has been identified between belinostat glucuronide formation and glucuronidation of known UGT1A1 substrates. Belinostat glucuronide 124-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 23382909-7 2013 The main metabolic pathway of belinostat is through glucuronidation mediated primarily by UGT1A1, a highly polymorphic enzyme. belinostat 30-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 23236239-0 2012 UGT1A1 predicts outcome in colorectal cancer treated with irinotecan and fluorouracil. Fluorouracil 73-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23236239-9 2012 Differences in irinotecan and its metabolites between UGT1A1 gene variants were compared. Irinotecan 15-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 23236239-21 2012 Irinotecan PK was investigated in 34 patients, which showed high area under concentration curve (AUC) of irinotecan and SN-38, but low AUC ratio (SN-38G / SN-38) in those patients with UGT1A1*28 TA7/7. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 23236239-22 2012 CONCLUSION: A distinct distribution pattern of UGT1A1 genotypes in Chinese patients might contribute to relatively low toxicity associated with irinotecan and 5-fluorouracil in mCRC patients. Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 23236239-22 2012 CONCLUSION: A distinct distribution pattern of UGT1A1 genotypes in Chinese patients might contribute to relatively low toxicity associated with irinotecan and 5-fluorouracil in mCRC patients. Fluorouracil 159-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 23279026-4 2012 It is well-known that UGT1A1, UGT1A6 and UGT1A9 enzymes glucuronidate acetaminophen. Acetaminophen 70-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). glycitein 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Sulfates 62-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). glycitein 220-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Isoflavones 277-288 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Sulfates 292-300 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Sulfates 292-300 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Glucuronides 339-350 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-5 2012 UGT1A1*28 was associated only with percentage of glycitein as sulphate (positive; p = 0.046), but excluding five participants with both minor alleles of CBG and ABCG2 uncovered additional associations with percentage of glycitein as glucuronide (negative; p = 0.028), combined isoflavones as sulphate (positive; p = 0.035) and sulphate-to-glucuronide ratio for combined isoflavones (positive; p = 0.036). Isoflavones 370-381 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 23097198-7 2012 CONCLUSION: Genetic variation in UGT1A1, CBG, ABCG2, and ABCC2 influences isoflavone metabolism so may affect benefits of dietary consumption. Isoflavones 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 23207817-1 2012 Bilirubin is conjugated with glucoronic acid in the liver by UDP-glucuronosyltransferase 1A1 (UGT1A1). Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-92 23233861-7 2012 Of the proposed gene/drug pairs the highest importance was assigned to HLA-B/abacavir, UGT1A1/irinotecan, and CYP2D6/tamoxifen. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 23207817-1 2012 Bilirubin is conjugated with glucoronic acid in the liver by UDP-glucuronosyltransferase 1A1 (UGT1A1). Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 23207817-1 2012 Bilirubin is conjugated with glucoronic acid in the liver by UDP-glucuronosyltransferase 1A1 (UGT1A1). glucoronic acid 29-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-92 23207817-1 2012 Bilirubin is conjugated with glucoronic acid in the liver by UDP-glucuronosyltransferase 1A1 (UGT1A1). glucoronic acid 29-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 22648071-0 2012 UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for puerarin metabolism in human liver microsomes. puerarin 72-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 22648071-6 2012 In vitro, we used a UDP-glucuronosyltransferase (UGT) reaction screening method with 12 recombinant human UGTs to demonstrate that formation of puerarin-7-O-glucuronide was catalyzed by UGT1A1, 1A9, 1A10, 1A3, 1A6, 1A7, and 1A8. puerarin-7-O-glucuronide 144-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 22648071-7 2012 UGT1A1, 1A9, and 1A10 significantly catalyzed puerarin-7-O-glucuronide formation, and the activity of UGT1A1 was significantly higher than those of 1A9 and 1A10. puerarin-7-O-glucuronide 46-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22648071-9 2012 The kinetics of puerarin-7-O-glucuronide formation catalyzed by UGT1A1 were similar to those of the pooled human liver microsomes (HLMs), with V (max) values of 186.3 and 149.2 pmol/min/mg protein, and K ( m ) values of 811.3 and 838.9 muM, respectively. puerarin-7-O-glucuronide 16-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22648071-10 2012 Furthermore, bilirubin and beta-estradiol, probe substrates for UGT1A1, significantly inhibited the formation of puerarin-7-O-glucuronide in HLMs. Bilirubin 13-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22648071-10 2012 Furthermore, bilirubin and beta-estradiol, probe substrates for UGT1A1, significantly inhibited the formation of puerarin-7-O-glucuronide in HLMs. Estradiol 27-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22648071-10 2012 Furthermore, bilirubin and beta-estradiol, probe substrates for UGT1A1, significantly inhibited the formation of puerarin-7-O-glucuronide in HLMs. puerarin-7-O-glucuronide 113-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22829544-1 2012 Human UDP-glucuronosyltransferase (UGT) 1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds such as bilirubin. Bilirubin 168-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 22829544-2 2012 The present study shows how cyclin-dependent kinase (CDK) inhibitor roscovitine stimulated the expression of UGT1A1 in HepG2 cells. Roscovitine 68-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 22829544-3 2012 Pregnane X receptor (PXR)-mediated transactivation of UGT1A1 reporter gene was more prominently enhanced by roscovitine, compared with the basal-, constitutive androstane receptor (CAR)-, and aryl hydrocarbon receptor-mediated activities. Roscovitine 108-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22829544-4 2012 We determined the regulatory mechanism of UGT1A1 expression through PXR"s stimulation by roscovitine. Roscovitine 89-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Roscovitine 151-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Rifampin 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22829544-5 2012 Although phosphomimetic mutations at Thr290 and Thr408 retained the PXR protein in cytoplasm and attenuated the induction of UGT1A1 expression by both roscovitine and rifampicin, a mutation at Ser350 specifically reduced the activity of PXR induced by roscovitine. Roscovitine 252-263 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22829544-10 2012 These results indicate that roscovitine stimulated the expression of UGT1A1 by inhibiting CDK2, which phosphorylated PXR at Ser350 to suppress binding with RXR and coactivator and maintain the acetylation of PXR protein. Roscovitine 28-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 22820246-2 2012 For examples: (i) bilirubin is solely conjugated by UGT1A1 and activates its transcription factors Ah receptor, PXR and CAR. Bilirubin 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 22212955-0 2012 Association of serum bilirubin and promoter variations in HMOX1 and UGT1A1 genes with sporadic colorectal cancer. Bilirubin 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 22212955-1 2012 Heme oxygenase-1 (HMOX1) and bilirubin UDP-glucuronosyltransferase (UGT1A1) enzymes, both involved in bilirubin homeostasis, play an important role in the oxidative stress defense. Bilirubin 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 22212955-4 2012 The (GT)(n) and (TA)(n) dinucleotide variations in HMOX1 and UGT1A1 gene promoters, respectively, were determined by fragment analysis. (n) dinucleotide 21-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 23323463-8 2012 The prodrug irinotecan is biotransformed by carboxylesterase into its active metabolite SN-38, which is inactivated by UGT1 into the inactive compound SN-38G. Irinotecan 151-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-123 22050734-3 2012 Some protease inhibitors (mainly atazanavir and indinavir) act as inhibitors of uridine diphosphate-glucuronosyltransferase (UGT1A1), the enzyme responsible for hepatic conjugation of bilirubin. Atazanavir Sulfate 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22910411-3 2012 Using HepG2 cells expressing human CAR in the presence of tetracycline, we showed that knockdown of DP97 with small interfering RNAs suppressed tetracycline-inducible mRNA expression of CYP2B6 and UGT1A1 but not CYP3A4. Tetracycline 144-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 197-203 22050734-3 2012 Some protease inhibitors (mainly atazanavir and indinavir) act as inhibitors of uridine diphosphate-glucuronosyltransferase (UGT1A1), the enzyme responsible for hepatic conjugation of bilirubin. Indinavir 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22050734-3 2012 Some protease inhibitors (mainly atazanavir and indinavir) act as inhibitors of uridine diphosphate-glucuronosyltransferase (UGT1A1), the enzyme responsible for hepatic conjugation of bilirubin. Bilirubin 184-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 22050734-4 2012 Variations in the promoter region of the UGT1A1 gene (UGT1A1*28, rs8175347) can influence bilirubin plasma levels, modulating the susceptibility to hyperbilirubinemia. Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 22050734-4 2012 Variations in the promoter region of the UGT1A1 gene (UGT1A1*28, rs8175347) can influence bilirubin plasma levels, modulating the susceptibility to hyperbilirubinemia. Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22050734-6 2012 Individuals carrying the UGT1A1*28 allele had a higher risk of developing severe hyperbilirubinemia [prevalence ratio (PR)=2.43, 95% confidence interval (CI) 1.08-5.45, p=0.032] as well as atazanavir users (PR=7.72, 95% CI=3.14-18.98, p<0.001). Atazanavir Sulfate 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 22050734-7 2012 This is the first description of such an association in Brazilian HIV patients, which shows that in African-American and Euroamerican HAART users, the UGT1A1*28 allele also predisposes to severe hyperbilirubinemia, especially in those exposed to atazanavir. Atazanavir Sulfate 246-256 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-157 22661571-5 2012 RESULTS: At baseline, UGT1A1 heterozygous and homozygous patients had significantly higher bilirubin levels than wild-type (P = 0.012 and P < 0.001, respectively). Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 22661571-9 2012 CONCLUSIONS: UGT1A1*28 is significantly related to hyperbilirubinaemia in HIV-1 patients receiving atazanavir. Atazanavir Sulfate 99-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 22661571-11 2012 Unboosted atazanavir-containing therapy is safe and efficacious in patients with an undetectable viral load with a UGT1A1*28 polymorphism, allowing the use of atazanavir in patients otherwise likely unable to receive it. Atazanavir Sulfate 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 22661630-6 2012 The expression of UGT1A1 v2/v3 was 1.6-fold higher than v1 (p = 0.03) in HepG2 cells, and short interfering RNA knockdown of HepG2 v2/v3 increased raloxifene glucuronidation activity by 83%. Raloxifene Hydrochloride 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 22435749-3 2012 Unique tryptic peptides of UGT1A1 and UGT2B7 in tryptically digested HLMs were simultaneously quantified by liquid chromatography (LC) equipped with tandem mass spectrometry (MS) using corresponding stable isotope-labelled peptides as internal standards. Peptides 15-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 23016456-4 2012 Among the tested UGT isoforms, UGT1A1, UGT1A3, UGT2B7 and UGT2B15 were strongly inhibited by tacro in a concentration-dependent manner. Tacrolimus 93-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 22435749-3 2012 Unique tryptic peptides of UGT1A1 and UGT2B7 in tryptically digested HLMs were simultaneously quantified by liquid chromatography (LC) equipped with tandem mass spectrometry (MS) using corresponding stable isotope-labelled peptides as internal standards. Peptides 223-231 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 22749334-0 2012 Effects of statin treatments and polymorphisms in UGT1A1 and SLCO1B1 on serum bilirubin levels in Chinese patients with hypercholesterolaemia. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 21978357-1 2012 Raltegravir is metabolized by glucuronidation via UDP-glucuronosyltransferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-81 21978357-1 2012 Raltegravir is metabolized by glucuronidation via UDP-glucuronosyltransferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 21978357-2 2012 We analyzed the genotypes of UGT1A1 (*6, *27, and *28) and their contribution to plasma raltegravir concentrations in 56 Japanese HIV-1-infected patients in the National Hospital Organization Nagoya Medical Center of Japan. Raltegravir Potassium 88-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 21978357-9 2012 However, variability in raltegravir concentration and small patient population precluded a correlation between UGT1A1*6 homozygosity and plasma raltegravir concentration. Raltegravir Potassium 144-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 21978357-10 2012 To clarify the contribution of UGT1A1*6 or *28 polymorphisms to plasma raltegravir concentrations, further investigations on larger subject populations are required. Raltegravir Potassium 71-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 22749334-2 2012 The organic anion transporting polypeptide 1B1 (OATP1B1, gene SLCO1B1) and UDP-glucuronosyltransferase 1A1 (UGT1A1) play an important role in the disposition of bilirubin. Bilirubin 161-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-106 22749334-2 2012 The organic anion transporting polypeptide 1B1 (OATP1B1, gene SLCO1B1) and UDP-glucuronosyltransferase 1A1 (UGT1A1) play an important role in the disposition of bilirubin. Bilirubin 161-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 22749334-4 2012 METHODS: Associations between common polymorphisms in UGT1A1 and SLCO1B1 and the serum bilirubin levels on no lipid-lowering treatment were analyzed in 379 Chinese patients with hypercholesterolaemia. Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22749334-6 2012 RESULTS: The UGT1A1 polymorphisms associated with reduced enzyme activity were significantly associated with increased baseline bilirubin levels. Bilirubin 128-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 22749334-9 2012 CONCLUSIONS: This study showed that the polymorphisms in UGT1A1, but not SLCO1B1, were associated with serum bilirubin levels in Chinese patients. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 22943620-4 2012 In a recombinant human UGT1A1 assay, the assay buffer components uridine and its phosphorylated derivatives are isobaric with RBV and its phosphorylated metabolites, leading to significant interference when analyzed by LC-MS with the nominal mass resolution mode. Uridine 65-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 22943620-4 2012 In a recombinant human UGT1A1 assay, the assay buffer components uridine and its phosphorylated derivatives are isobaric with RBV and its phosphorylated metabolites, leading to significant interference when analyzed by LC-MS with the nominal mass resolution mode. Ribavirin 126-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 22943620-6 2012 CONCLUSION: The developed LC-high-resolution accurate MS method allows for quantitation of RBV and its phosphorylated metabolites, eliminating the interferences from uridine and its phosphorylated derivatives in recombinant human UGT1A1 assays. Ribavirin 91-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 22960892-1 2012 BACKGROUND & OBJECTIVES: Genetic polymorphisms of uridine diphosphate glucuronyltransferase 1A1 (UGT1A1) have been associated with a wide variation of responses among patients prescribed with irinotecan. Adenosine Monophosphate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-1 2012 BACKGROUND & OBJECTIVES: Genetic polymorphisms of uridine diphosphate glucuronyltransferase 1A1 (UGT1A1) have been associated with a wide variation of responses among patients prescribed with irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-3 2012 The objective of this study was to investigate the prevalence of three different variants of UGT1A1 (UGT1A1*6, UGT1A1*27 and UGT1A1*28), which are associated with reduced enzyme activity and increased irinotecan toxicity, in the three main ethnic groups in Malaysia (Malays, Chinese and Indians). Irinotecan 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22960892-11 2012 INTERPRETATION & CONCLUSIONS: There was interethnic variability in the frequency of UGT1A1*28 in the Malaysian population. Adenosine Monophosphate 16-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 22960892-12 2012 Our results suggest that genotyping of UUGT1A1*6, UGT1A1*28 and UGT1A1*27 need to be performed before patients are prescribed with irinotecan due to their high prevalence of allelic variant which could lead to adverse drug reaction. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22565219-7 2012 UGT1A1 showed a 2.1-fold (P <0.0001) higher V(max)/K(M) for formation of the CLZ-N+-glucuronide than UGT1A4; UGT1A4 was the only UGT for which CLZ-5-N-glucuronide kinetics could be determined. clozapine N-glucuronide 80-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22957438-3 2012 The results showed that 100 microM of glimepiride inhibited UGT1A1, UGT1A3, UGT1A6, UGT1A9, UGT2B7 and UGT2B15 by 54.7%, 43.1%, 100%, 70.5%, 32.7 and 37.2%, respectively. glimepiride 38-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 22565219-7 2012 UGT1A1 showed a 2.1-fold (P <0.0001) higher V(max)/K(M) for formation of the CLZ-N+-glucuronide than UGT1A4; UGT1A4 was the only UGT for which CLZ-5-N-glucuronide kinetics could be determined. clz-5-n-glucuronide 146-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22565219-10 2012 The UGT1A1*28 allele was a significant (P = 0.045) predictor of CLZ-N+-glucuronide formation; the UGT1A4*3 allele was a significant (P < 0.0001) predictor of CLZ-5-N-glucuronide and dmCLZ-glucuronide formation. clozapine N-glucuronide 64-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 22565219-11 2012 CONCLUSION: These data suggest that the UGT1A1*28 and UGT1A4*3 alleles contribute significantly to the interindividual variability in CLZ and dmCLZ metabolism. Clozapine 134-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22565219-11 2012 CONCLUSION: These data suggest that the UGT1A1*28 and UGT1A4*3 alleles contribute significantly to the interindividual variability in CLZ and dmCLZ metabolism. norclozapine 142-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22367021-12 2012 However, the predicted high activity UGT1A1 genotype, associated with low serum levels of the antioxidant bilirubin, was associated with an increased ESCC risk. Bilirubin 106-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 248-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 248-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 248-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 22398043-3 2012 UDP-glucuronosyltransferase 1A1 (UGT1A1) is the only enzyme involved in the conjugation of bilirubin and the common UGT1A1*28 allele in the UGT1A1 gene, which is strongly associated with Gilbert"s syndrome in Caucasians, results in elevated plasma bilirubin levels. Bilirubin 248-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 22398043-10 2012 CONCLUSION: The homozygous state of the UGT1A1*28 polymorphism, associated with higher serum bilirubin levels, may be protective for the development of Crohn"s disease, suggesting that the anti-oxidant capacity of bilirubin may play a part. Bilirubin 93-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22398043-10 2012 CONCLUSION: The homozygous state of the UGT1A1*28 polymorphism, associated with higher serum bilirubin levels, may be protective for the development of Crohn"s disease, suggesting that the anti-oxidant capacity of bilirubin may play a part. Bilirubin 214-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22676194-0 2012 Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 22676194-1 2012 AIM: Pre-emptive irinotecan dose reduction for UGT1A1*28 homozygotes may result in reduced risk of severe neutropenia and diarrhea. Irinotecan 17-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 22676194-3 2012 Whether UGT1A1*28 genotype is associated with irinotecan response therefore is an important gap in existing knowledge to inform clinical utility. Irinotecan 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 22676194-4 2012 MATERIALS & METHODS: A systematic review and meta-analysis was performed to analyze the difference in objective response rate (ORR) between irinotecan-administered cancer patients with different UGT1A1*28 genotypes: *28/*28 (homozygous variant), *1/*28 (heterozygous variant) or *1/*1 (wild-type). Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 22676194-5 2012 The effect of irinotecan dose on the association between UGT1A1*28 and ORR was also assessed. Irinotecan 14-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 22516947-0 2012 Phase II and UGT1A1 genotype study of irinotecan dose escalation as salvage therapy for advanced gastric cancer. Irinotecan 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 22170007-1 2012 PURPOSE: Axitinib, an orally administered inhibitor of vascular endothelial growth factor 1, 2 and 3, is primarily metabolized by cytochrome P450 (CYP) 3A4/5 but is also a substrate for CYP1A2, CYP2C19, UDP-glucuronosyltransferase (UGT)1A1 and the drug transporters P-glycoprotein (encoded by the ABCB1 gene) and OATP1B1 (encoded by SLC01B1). Axitinib 9-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 232-239 22427354-7 2012 Epithelial markers for cell proliferation (Ki-67), terminal differentiation (transglutaminase 1-TGase1) and glucuronidation (UDP-glucuronosyltransferase1A1-UGT1A1) exhibited fenretinide concentration-specific relationships (elevated TGase1 and UGT1A1 levels <5 muM, reduced Ki-67 indices >5 muM) relative to blank-treated epithelium. Fenretinide 174-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-155 22321050-1 2012 We recently designed the CIME cocktail consisting of 10 drugs to assess the activity of the major human CYPs (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A), a phase II enzyme (UGT1A1/6/9), two drug transporters (P-gp and OATP1B1) and a component of the renal function ( Videau et al. cime 25-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-191 22543504-0 2012 Gossypol exhibits a strong influence towards UDP-glucuronosyltransferase (UGT) 1A1, 1A9 and 2B7-mediated metabolism of xenobiotics and endogenous substances. Gossypol 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-82 22543504-3 2012 In the present study, in vitro human liver microsomal incubation system supplemented with UDPGA was used to investigate the inhibition of gossypol towards UGT1A1, 1A9 and 2B7-mediated metabolism of xenobiotics and endogenous substances. Gossypol 138-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 22543504-4 2012 Estradiol, the probe substrate of UGT1A1, was selected as representative endogenous substance. Estradiol 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 22448797-2 2012 A polymorphism of the 5 end of the UGT1A1 gene promoter, a homozygous insertion of TA pairs (genotype UGT1A1*28/*28), results in a decrease in bilirubin glucuronidation activity and therefore leads to an increase in the level of unconjugated bilirubin (hyperbilirubinemia). Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22448797-2 2012 A polymorphism of the 5 end of the UGT1A1 gene promoter, a homozygous insertion of TA pairs (genotype UGT1A1*28/*28), results in a decrease in bilirubin glucuronidation activity and therefore leads to an increase in the level of unconjugated bilirubin (hyperbilirubinemia). Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22448797-2 2012 A polymorphism of the 5 end of the UGT1A1 gene promoter, a homozygous insertion of TA pairs (genotype UGT1A1*28/*28), results in a decrease in bilirubin glucuronidation activity and therefore leads to an increase in the level of unconjugated bilirubin (hyperbilirubinemia). Bilirubin 243-252 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22448797-2 2012 A polymorphism of the 5 end of the UGT1A1 gene promoter, a homozygous insertion of TA pairs (genotype UGT1A1*28/*28), results in a decrease in bilirubin glucuronidation activity and therefore leads to an increase in the level of unconjugated bilirubin (hyperbilirubinemia). Bilirubin 243-252 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22307138-11 2012 CONCLUSIONS: This pilot study indicates that genotype status of UGT1A1, UGT1A9, and ABCC2 and serum bilirubin concentration increases reflect abnormally high AUC in patients treated with sorafenib. Sorafenib 187-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22357454-2 2012 Lapatinib-associated Hy"s Law cases have been characterized using human leukocyte antigen (HLA) DQA1*02:01/DRB1*07:01 and Gilbert"s syndrome UGT1A1*28/*28 genotypes. Lapatinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 22085899-0 2012 UGT1A1 is a major locus influencing bilirubin levels in African Americans. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22085899-11 2012 In summary, UGT1A1 is a major locus influencing bilirubin levels and the results of this study promise to contribute to understanding of the etiology and treatment of hyperbilirubinaemia in African-ancestry populations. Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 22537951-9 2012 The Arg allele frequency of the polymorphisms of UGT1A1 gene in the case group was significantly higher than in the control group (P<0.01). Arginine 4-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 22307138-0 2012 Sorafenib is an inhibitor of UGT1A1 but is metabolized by UGT1A9: implications of genetic variants on pharmacokinetics and hyperbilirubinemia. Sorafenib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 22307138-1 2012 PURPOSE: Several case reports suggest sorafenib exposure and sorafenib-induced hyperbilirubinemia may be related to a (TA)(5/6/7) repeat polymorphism in UGT1A1*28 (UGT, uridine glucuronosyl transferase). Sorafenib 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 22307138-1 2012 PURPOSE: Several case reports suggest sorafenib exposure and sorafenib-induced hyperbilirubinemia may be related to a (TA)(5/6/7) repeat polymorphism in UGT1A1*28 (UGT, uridine glucuronosyl transferase). Sorafenib 61-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 22307138-2 2012 We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Sorafenib 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 22307138-2 2012 We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Sorafenib 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 22307138-2 2012 We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Sorafenib 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 22307138-2 2012 We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Sorafenib 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 22307138-2 2012 We hypothesized that sorafenib inhibits UGT1A1 and individuals carrying UGT1A1*28 and/or UGT1A9 variants experience greater sorafenib exposure and greater increase in sorafenib-induced plasma bilirubin concentration. Bilirubin 192-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 22307138-3 2012 EXPERIMENTAL DESIGN: Inhibition of UGT1A1-mediated bilirubin glucuronidation by sorafenib was assessed in vitro. Bilirubin 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 22307138-3 2012 EXPERIMENTAL DESIGN: Inhibition of UGT1A1-mediated bilirubin glucuronidation by sorafenib was assessed in vitro. Sorafenib 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 22307138-6 2012 RESULTS: Sorafenib exhibited mixed-mode inhibition of UGT1A1-mediated bilirubin glucuronidation (IC(50) = 18 mumol/L; K(i) = 11.7 mumol/L) in vitro. Sorafenib 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22307138-6 2012 RESULTS: Sorafenib exhibited mixed-mode inhibition of UGT1A1-mediated bilirubin glucuronidation (IC(50) = 18 mumol/L; K(i) = 11.7 mumol/L) in vitro. Bilirubin 70-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22307138-9 2012 Sorafenib exposure was related to plasma bilirubin increases in patients carrying 1 or 2 copies of UGT1A1*28 alleles (n = 12 and n = 5; R(2) = 0.38 and R(2) = 0.77; P = 0.032 and P = 0.051, respectively). Sorafenib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 22307138-10 2012 UGT1A1*28 carriers showed two distinct phenotypes that could be explained by ABCC2-24C>T genotype and are more likely to experience plasma bilirubin increases following sorafenib if they had high sorafenib exposure. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22307138-10 2012 UGT1A1*28 carriers showed two distinct phenotypes that could be explained by ABCC2-24C>T genotype and are more likely to experience plasma bilirubin increases following sorafenib if they had high sorafenib exposure. Sorafenib 172-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22307138-10 2012 UGT1A1*28 carriers showed two distinct phenotypes that could be explained by ABCC2-24C>T genotype and are more likely to experience plasma bilirubin increases following sorafenib if they had high sorafenib exposure. Sorafenib 199-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 22021027-6 2012 Samples with higher concentrations of n-6 fatty acids produced a higher expression of UGT1A1 mRNA. Fatty Acids, Omega-6 38-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 22416852-7 2012 Study subjects were genotyped at the UGT1A1*28 locus, which is strongly associated with bilirubin levels. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 21923755-4 2012 KEY RESULTS: Intracellular accumulation of ezetimibe was significantly lower in MDCK-OATP1B1-UGT1A1-MRP2 triple-transfected cells compared with all other cell lines. Ezetimibe 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21923755-5 2012 Considerably higher amounts of ezetimibe glucuronide were found in the apical compartment of MDCK-OATP1B1-UGT1A1-MRP2 monolayers compared with all other cell lines. ezetimibe glucuronide 31-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 21923755-7 2012 Intracellular concentrations of etoposide equivalents (i.e. parent compound plus metabolites) were affected only to a minor extent by the absence or presence of OATP1B1/UGT1A1/MRP2. Etoposide 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 21923755-8 2012 In contrast, apical accumulation of etoposide equivalents was significantly higher in monolayers of both cell lines expressing MRP2 (MDCK-OATP1B1-MRP2, MDCK-OATP1B1-UGT1A1-MRP2) compared with the single-transfected (OATP1B1) and the control cell line. Etoposide 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 22180045-5 2012 Assays with recombinant human UDP-glucuronosyltransferase enzymes (UGTs) revealed that multiple UGT isoforms were involved in magnolol glucuronidation, including UGT1A1, -1A3, -1A7, -1A8, -1A9, -1A10, and -2B7. magnolol 126-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 22297387-7 2012 Efavirenz (single dose) slightly increased ezetimibe absorption and markedly decreased exposure to ezetimibe-glucuronide (single dose and multiple dose), which may be explained by inhibition of UGT1A1 and ABCB1 (in vitro data). efavirenz 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 22297387-7 2012 Efavirenz (single dose) slightly increased ezetimibe absorption and markedly decreased exposure to ezetimibe-glucuronide (single dose and multiple dose), which may be explained by inhibition of UGT1A1 and ABCB1 (in vitro data). ezetimibe glucuronide 99-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 22154984-0 2012 Evaluation of bisphenol A glucuronidation according to UGT1A1*28 polymorphism by a new LC-MS/MS assay. bisphenol A 14-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 22740978-0 2012 Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. 7-ethyl-10-hydroxycamptothecin glucuronide 51-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 22740978-0 2012 Correlation between plasma concentration ratios of SN-38 glucuronide and SN-38 and neutropenia induction in patients with colorectal cancer and wild-type UGT1A1 gene. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 22740978-3 2012 A total of 17 patients with colorectal cancer and wild-type UDP-glucuronosyltransferase (UGT)1A1 gene were enrolled and treated with CPT-11 as part of the FOLFIRI regimen [CPT-11 and fluorouracil (5-FU)]. Irinotecan 133-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-96 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22740978-9 2012 In conclusion, UGT activity involved in SN-38 metabolism is variable among patients with the wild-type UGT1A1 gene, and each CPT-11 treatment suppresses UGT activity. Irinotecan 125-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 22347861-3 2012 Protection from cardiovascular disease is also observed in patients with Gilbert"s syndrome which is a disease characterized by mutations in hepatic UGT1A1, the enzyme responsible for the conjugation of bilirubin into the bile. Bilirubin 203-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 6-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 6-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 97-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 97-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 97-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 22154984-8 2012 Since BPA is a UGT1A1 substrate, we postulated that the common UGT1A1*28 polymorphism influences BPA glucuronidation, and consequently, BPA detoxification. bisphenol A 97-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 21760472-0 2012 Bilirubin dependence on UGT1A1 polymorphisms, hemoglobin, fasting time and body mass index. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21760472-2 2012 This study evaluates how several nongenetic causes and the genetic UGT1A1 polymorphisms contribute for bilirubin levels, in a cohort of 146 young Caucasian females. Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 21953915-6 2012 Among the tested human UGT isoforms, UGT1A7, UGT1A8, and UGT1A9 showed the highest activity for the conjugation of hydroxylated TCC metabolites followed by UGT1A1, UGT1A3, and UGT1A10. triclocarban 128-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-162 22252501-6 2012 Efavirenz also moderately inhibited UGT1A1-mediated 17beta-estradiol 3-glucuronidation, with a K(i) value of 40.3 muM, but did not inhibit UGT1A6-mediated 1-naphthol glucuronidation. efavirenz 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22252501-6 2012 Efavirenz also moderately inhibited UGT1A1-mediated 17beta-estradiol 3-glucuronidation, with a K(i) value of 40.3 muM, but did not inhibit UGT1A6-mediated 1-naphthol glucuronidation. Estradiol 52-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22252501-6 2012 Efavirenz also moderately inhibited UGT1A1-mediated 17beta-estradiol 3-glucuronidation, with a K(i) value of 40.3 muM, but did not inhibit UGT1A6-mediated 1-naphthol glucuronidation. 1-naphthol 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 22293344-4 2012 The aim of this study was to clarify the mechanism of these phenomena in HLM and human intestinal microsomes (HIM) and its relevance to uridine 5"-diphosphate (UDP)-glucuronosyl transferase (UGT) 1A1, UGT2B4 and UGT2B7, which mainly metabolize carvedilol directly in phase II enzymes. Carvedilol 244-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-199 22201120-2 2012 All patients harbored UDP-glucuronosyltransferase (UGT) 1A1*1/*1, *1/*6, or *1/*28 genotypes, which are associated with similar irinotecan pharmacokinetics and responses to FOLFIRI. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-59 22118420-0 2012 A genome-wide search for non-UGT1A1 markers associated with unconjugated bilirubin level reveals significant association with a polymorphic marker near a gene of the nucleoporin family. Bilirubin 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 22118420-1 2012 Variants in the UGT1A1 gene and its promoter are known to determine levels of unconjugated bilirubin (UCB), but do not explain all cases of unconjugated hyperbilirubinemia. Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 22118420-1 2012 Variants in the UGT1A1 gene and its promoter are known to determine levels of unconjugated bilirubin (UCB), but do not explain all cases of unconjugated hyperbilirubinemia. ucb 102-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 22447115-4 2012 When cells were treated with a typical CYP3A substrate (triazolam), CYP2C9 substrate (diclofenac) or UGT1A1 substrate (SN-38), large amounts of their metabolites were detected in the medium of cells cultured on micro-space cell culture plates. Irinotecan 119-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Triazolam 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22201120-2 2012 All patients harbored UDP-glucuronosyltransferase (UGT) 1A1*1/*1, *1/*6, or *1/*28 genotypes, which are associated with similar irinotecan pharmacokinetics and responses to FOLFIRI. FOLFIRI regimen 173-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-59 22673034-5 2012 The levels of CYP1A1, CYP1A2, and UGT1A1 mRNA expression were increased by omeprazole and 3-methylcholanthrene. Omeprazole 75-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 22572482-4 2012 Among the 12 human UGTs investigated, UGT1A1 and UGT1A9 were the major isoforms that catalyzed the glucuronidation of Sal A (K(m) values of 29.72 +- 2.20 and 24.40 +- 2.60 microM). salvianolic acid A 118-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Diclofenac 45-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Ketoconazole 129-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Sulfaphenazole 162-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Ketoconazole 200-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-188 22572482-8 2012 Mefenamic acid inhibited Sal A glucuronidation in UGT1A1 and HLMs with IC(50) values of >200 and 12.4 +- 2.2 microM, respectively. Mefenamic Acid 0-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 22673034-5 2012 The levels of CYP1A1, CYP1A2, and UGT1A1 mRNA expression were increased by omeprazole and 3-methylcholanthrene. Methylcholanthrene 90-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 22572482-8 2012 Mefenamic acid inhibited Sal A glucuronidation in UGT1A1 and HLMs with IC(50) values of >200 and 12.4 +- 2.2 microM, respectively. sal 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 22450351-0 2012 Non tumoral hyperserotoninaemia responsive to octreotide due to dual polymorphism in UGT1A1 and UGT1A6. Octreotide 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 22548118-3 2012 DA-9701 and Corydalis tuber extract slightly inhibited UGT1A1-mediated etoposide glucuronidation, with 50% inhibitory concentration (IC(50)) values of 188 and 290 mug/mL, respectively. etoposide glucuronidation 71-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 23964438-4 2012 It was demonstrated that the genetic variations cause a decrease in UGT1A1 activity in neonates, leading to an accumulation of unconjugated bilirubin in serum. Bilirubin 140-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 22567180-3 2012 METHODS: A novel oligonucleotide microarray was developed to genotype 13 variations (DPYD*2, DPYD*5, DPYD*9, TYMS 6 bp Ins/Del, UGT1A1*6, UGT1A1*27, UGT1A1*28, GSTP1 Ile105Val, XRCC1 Arg399Gln, MTHFR C677T, MDR1 C3435T/A, MDR1 G2677A/T and ERCC1 C118T). Oligonucleotides 17-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 22567180-3 2012 METHODS: A novel oligonucleotide microarray was developed to genotype 13 variations (DPYD*2, DPYD*5, DPYD*9, TYMS 6 bp Ins/Del, UGT1A1*6, UGT1A1*27, UGT1A1*28, GSTP1 Ile105Val, XRCC1 Arg399Gln, MTHFR C677T, MDR1 C3435T/A, MDR1 G2677A/T and ERCC1 C118T). Oligonucleotides 17-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 22567180-3 2012 METHODS: A novel oligonucleotide microarray was developed to genotype 13 variations (DPYD*2, DPYD*5, DPYD*9, TYMS 6 bp Ins/Del, UGT1A1*6, UGT1A1*27, UGT1A1*28, GSTP1 Ile105Val, XRCC1 Arg399Gln, MTHFR C677T, MDR1 C3435T/A, MDR1 G2677A/T and ERCC1 C118T). Oligonucleotides 17-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 22508373-0 2012 A phase I study of infusional 5-fluorouracil, leucovorin, oxaliplatin and irinotecan in Japanese patients with advanced colorectal cancer who harbor UGT1A1*1/*1,*1/*6 or *1/*28. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 23430851-2 2012 It is caused by deficiency of the liver enzyme responsible for bilirubin elimination, the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1; EC 2.4.1.17). Bilirubin 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-137 23430851-2 2012 It is caused by deficiency of the liver enzyme responsible for bilirubin elimination, the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1; EC 2.4.1.17). Bilirubin 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 21972237-6 2012 In particular, UGT1A1 and extrahepatic UGT1A10 have significantly higher capacities than other isoforms for S-7-hydroxywarfarin and R-7-hydroxywarfarin glucuronidation, respectively. 7-hydroxywarfarin 108-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21972237-6 2012 In particular, UGT1A1 and extrahepatic UGT1A10 have significantly higher capacities than other isoforms for S-7-hydroxywarfarin and R-7-hydroxywarfarin glucuronidation, respectively. (R)-7-Hydroxywarfarin 132-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21972237-7 2012 Activity data generated using a set of well characterized human liver microsomes supported the recombinant enzyme data, suggesting an important (although not exclusive) role for UGT1A1 in glucuronidation of the main warfarin metabolites, including Rac-6- and 7-hydroxywarfarin and their R- and S-enantiomers in the liver. Warfarin 216-224 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-184 21972237-8 2012 This is the first demonstration that the R- and S-enantiomers of hydroxywarfarins are glucuronidated, with significantly different enzymatic affinity and capacity, and supports the importance of UGT1A1 as the major hepatic isoform involved. hydroxywarfarins 65-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 195-201 21937736-7 2011 The aim of our in vitro study was to explain the mechanism behind the observed influence of UGT1A1*28 polymorphism on raloxifene pharmacokinetics in a small-sized in vivo study (Br J Clin Pharmacol 67:437-444, 2009). Raloxifene Hydrochloride 118-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 22558097-1 2012 Serum bilirubin levels have been associated with polymorphisms in the UGT1A1 promoter in normal populations and in patients with hemolytic anemias, including sickle cell anemia. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 22293705-0 2012 [Examination of factors affecting adverse reactions and dosage reduction in UGT1A1 genotyped patients: a retrospective survey of irinotecan]. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 22293705-1 2012 Our aim was to clarify the side effects of irinotecan which occurred in patients admitted to Showa University Hospital to investigate whether the UGT1A1 genetic polymorphism status was reflected in the discontinuation or dose reduction of irinotecan. Irinotecan 239-249 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 22293705-8 2012 Our investigation confirmed that the UGT1A1 genetic polymorphism status of the patients was reflected in the discontinuance or dose reduction of irinotecan. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 21985641-5 2011 UGT1A1 activity correlation analyses using flavonoids-4"-O-glucuronidation vs beta-estradiol-3-glucuronidation (a well-recognized marker for UGT1A1) or vs SN-38 glucuronidation were performed using a bank of HLMs (n = 12) including three UGT1A1-genotyped HLMs (i.e., UGT1A1*1*1, UGT1A1*1*28, and UGT1A1*28*28). Irinotecan 155-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. Flavonoids 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-38 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. Flavonoids 108-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 4"-o 173-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-38 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 4"-o 173-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3',4'-dihydroxyflavonol 197-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-38 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3',4'-dihydroxyflavonol 197-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3,6,4"-thf 213-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-38 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3,6,4"-thf 213-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3-o 276-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-38 21985641-6 2011 The results showed that UGT1A1 and 1A9, followed by 1A7, were the main isoforms for glucuronidating the two flavonoids, where UGT1A1 accounted for 92 +- 7% and 91 +- 10% of 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF, respectively, and UGT1A9 accounted for most of the 3-O-glucuronidation. 3-o 276-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. 4"-o 31-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. 3',4'-dihydroxyflavonol 55-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. 3,6,4"-thf 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. Estradiol 132-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. 3-oh 143-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-8 2011 In conclusion, UGT1A1-mediated 4"-O-glucuronidation of 3,3",4"-THF and 3,6,4"-THF was highly correlated with the glucuronidation of estradiol (3-OH) and SN-38. Irinotecan 153-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 21985641-9 2011 This study demonstrated for the first time that regioselective glucuronidation of flavonoids can be applied to probe hepatic UGT1A1 activity in vitro. Flavonoids 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 21726413-7 2011 The V(max) values for beta-estradiol of p.P364L-UGT1A1, 1A3, 1A7, 1A8 and 1A10 were 36.6%, 82.1%, 26.8%, 29.2% and 22.5%, respectively, of the corresponding wild-type. Estradiol 23-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 21726413-8 2011 Glucuronidation activity towards acetaminophen of p.P364L-UGT1A1, 1A6, 1A7, 1A8, 1A9 and 1A10 was 50.3%, 46.4%, 17.2%, 44.1%, 5.0% and 42.8%, respectively, of wild-type. Acetaminophen 33-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 21787264-5 2011 In this view, the uridine diphosphate glucuronosil transferase isoform 1A1 (UGT1A1) was associated with significant changes in disposition of CPT-11 and its metabolites, and consequently with treatment-induced toxicities. Irinotecan 142-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-74 21787264-5 2011 In this view, the uridine diphosphate glucuronosil transferase isoform 1A1 (UGT1A1) was associated with significant changes in disposition of CPT-11 and its metabolites, and consequently with treatment-induced toxicities. Irinotecan 142-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 22077505-3 2011 METHODS: Published papers and publicly accessible genomic databases were searched up to August 2009 to obtain allele and genotype frequencies for UGT1A1*6 and *28, two common variants of UGT1A1, a gene that encodes for a key enzyme in the pathway of irinotecan metabolism. Irinotecan 250-260 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 22077505-3 2011 METHODS: Published papers and publicly accessible genomic databases were searched up to August 2009 to obtain allele and genotype frequencies for UGT1A1*6 and *28, two common variants of UGT1A1, a gene that encodes for a key enzyme in the pathway of irinotecan metabolism. Irinotecan 250-260 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 22077505-6 2011 Indians are at an elevated risk of irinotecan-induced neutropenia associated with UGT1A1*28 compared with Chinese and Japanese, and at an even higher risk compared with North American Caucasians. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 22077505-7 2011 On the other hand, Chinese and Japanese are at an elevated risk of irinotecan-induced neutropenia associated with UGT1A1*6 relative to Indians in Singapore or North American Caucasians. Irinotecan 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 21985641-2 2011 Here, regioselective glucuronidation of two flavonoids, 3,3",4"-trihydroxyflavone (3,3",4"-THF) and 3,6,4"-trihydroxyflavone (3,6,4"-THF), is used to probe the activity of hepatic UGT1A1. Flavonoids 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 21985641-2 2011 Here, regioselective glucuronidation of two flavonoids, 3,3",4"-trihydroxyflavone (3,3",4"-THF) and 3,6,4"-trihydroxyflavone (3,6,4"-THF), is used to probe the activity of hepatic UGT1A1. 3',4'-dihydroxyflavonol 56-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 21985641-2 2011 Here, regioselective glucuronidation of two flavonoids, 3,3",4"-trihydroxyflavone (3,3",4"-THF) and 3,6,4"-trihydroxyflavone (3,6,4"-THF), is used to probe the activity of hepatic UGT1A1. 3',4'-dihydroxyflavonol 83-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 21985641-2 2011 Here, regioselective glucuronidation of two flavonoids, 3,3",4"-trihydroxyflavone (3,3",4"-THF) and 3,6,4"-trihydroxyflavone (3,6,4"-THF), is used to probe the activity of hepatic UGT1A1. 3,6,4'-trihydroxyflavone 100-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 21985641-5 2011 UGT1A1 activity correlation analyses using flavonoids-4"-O-glucuronidation vs beta-estradiol-3-glucuronidation (a well-recognized marker for UGT1A1) or vs SN-38 glucuronidation were performed using a bank of HLMs (n = 12) including three UGT1A1-genotyped HLMs (i.e., UGT1A1*1*1, UGT1A1*1*28, and UGT1A1*28*28). Flavonoids 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21985641-5 2011 UGT1A1 activity correlation analyses using flavonoids-4"-O-glucuronidation vs beta-estradiol-3-glucuronidation (a well-recognized marker for UGT1A1) or vs SN-38 glucuronidation were performed using a bank of HLMs (n = 12) including three UGT1A1-genotyped HLMs (i.e., UGT1A1*1*1, UGT1A1*1*28, and UGT1A1*28*28). 4"-o 54-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21985641-5 2011 UGT1A1 activity correlation analyses using flavonoids-4"-O-glucuronidation vs beta-estradiol-3-glucuronidation (a well-recognized marker for UGT1A1) or vs SN-38 glucuronidation were performed using a bank of HLMs (n = 12) including three UGT1A1-genotyped HLMs (i.e., UGT1A1*1*1, UGT1A1*1*28, and UGT1A1*28*28). beta-estradiol-3 78-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-116 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). 7-ethyl-10-hydroxycamptothecin glucuronide 47-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-116 21840303-2 2011 SN-38 is metabolized to an inactive metabolite SN-38 glucuronide by uridine 5"-diphospho-glucuronosyltransferase 1A1 (UGT1A1). 7-ethyl-10-hydroxycamptothecin glucuronide 47-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 21840303-3 2011 Therefore, single nucleotide polymorphisms (SNPs) of the UGT1A1 gene are responsible for the severe adverse effects associated with the disruption of SN-38 metabolism. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 21840303-4 2011 However, despite having SNPs of the UGT1A1 gene, many patients metabolize SN-38 sufficiently to avoid severe adverse effects. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 35-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 21840303-9 2011 HGF suppressed the cytotoxicity of CPT-11 by decreasing intracellular SN-38 levels that resulted from a decrease in CES2 and an increase in UGT1A1. Irinotecan 70-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 21937736-8 2011 Incubation of raloxifene with human liver microsomes genotyped for UGT1A1*28 showed a significantly reduced metabolic clearance toward M1 in microsomes from donors with *28 allele. Raloxifene Hydrochloride 14-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 21937736-12 2011 Moreover, the significant influence of UGT1A1*28 polymorphism on metabolism of raloxifene was confirmed. Raloxifene Hydrochloride 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 21801714-2 2011 Free bilirubin is insoluble; its glucuronidation by bilirubin-UGT enzyme (UGT1A1) makes it soluble and eliminates it through urine and faeces. Bilirubin 5-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 21997136-8 2011 The EGFR inhibitors increased the S-phase and decreased the UGT1A1 and ABCG expression in irinotecan-resistant cells. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 22333276-0 2011 [Study of irinotecan-induced toxicity and its correlation to UGT1A1 gene promoter polymorphisms]. Irinotecan 10-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 22333276-1 2011 OBJECTIVES: To investigate the distribution of uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphism and its relation to the toxicities caused by irinotecan in Chinese patients with cervical cancer and ovarian cancer. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-94 22333276-1 2011 OBJECTIVES: To investigate the distribution of uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphism and its relation to the toxicities caused by irinotecan in Chinese patients with cervical cancer and ovarian cancer. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 22333276-5 2011 The relationship between UGT1A1 gene promoter polymorphism and the toxicity caused by irinotecan was analyzed. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 21801714-5 2011 Kinetic study of microsomal UGT1A1 from HepG2 cells suggested that Cln enhanced enzyme activity by increasing V(max) without altering K(m). carlinoside 67-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 21801714-7 2011 Since Nrf2 is the transcription factor of UGT1A1, we examined whether Cln effect on UGT1A1 overexpression is mediated through Nrf2. carlinoside 70-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 21801714-2 2011 Free bilirubin is insoluble; its glucuronidation by bilirubin-UGT enzyme (UGT1A1) makes it soluble and eliminates it through urine and faeces. Bilirubin 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 21801714-10 2011 Results from ChIP assay showed that Cln markedly augmented Nrf2 binding to UGT1A1 promoter that consequently enhanced reporter activity. carlinoside 36-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 21801714-11 2011 Our findings therefore show that Cln upregulated Nrf2 gene expression, increased its nuclear translocation and stimulated UGT1A1 promoter activity. carlinoside 33-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 21649640-4 2011 The aim of this study was to determine whether scoparone induces the expression of human UGT1A1. scoparone 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 21740478-0 2011 Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 21933112-6 2011 In particular, the structural basis for regioselective metabolism of flavonoids by SULT1A3 and UGT1A1 is discussed. Flavonoids 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 21892003-1 2011 Recent findings have disclosed the role of UDP-glucuronosyltransferase (UGT) 1A1*28 on the haematological toxicity induced by irinotecan (CPT-11), a drug commonly used in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-80 21892003-1 2011 Recent findings have disclosed the role of UDP-glucuronosyltransferase (UGT) 1A1*28 on the haematological toxicity induced by irinotecan (CPT-11), a drug commonly used in the treatment of metastatic colorectal cancer (mCRC). Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-80 21796020-2 2011 A polymorphic mutation (G71R) of bilirubin UDP-glucuronosyltransferase (UGT1A1) is a known cause of BMJ on the infantile side, but the responsible components of breast milk are not currently known. (12R)-12-methyltetradecanoic acid 100-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 21796020-3 2011 We analyzed the inhibitory effect of 5beta-pregnane-3alpha,20beta-diol (pregnanediol) on transcriptional activity and enzyme activity of UGT1A1. 5beta-pregnane-3alpha 37-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 21796020-3 2011 We analyzed the inhibitory effect of 5beta-pregnane-3alpha,20beta-diol (pregnanediol) on transcriptional activity and enzyme activity of UGT1A1. 20beta-diol 59-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 21796020-3 2011 We analyzed the inhibitory effect of 5beta-pregnane-3alpha,20beta-diol (pregnanediol) on transcriptional activity and enzyme activity of UGT1A1. Pregnanediol 72-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 21796020-8 2011 In contrast, in the presence of 100 muM pregnanediol, bilirubin glucuronidation of G71R-UGT1A1 was reduced to 51% of WT. Pregnanediol 40-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 21796020-8 2011 In contrast, in the presence of 100 muM pregnanediol, bilirubin glucuronidation of G71R-UGT1A1 was reduced to 51% of WT. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 21911884-3 2011 The (GT)n and (TA)n dinucleotide variations in HMOX1 and UGT1A1 gene promoters, respectively, were determined by fragment analysis in all subjects. Dinucleoside Phosphates 20-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 21740478-0 2011 Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21740478-1 2011 Irinotecan-induced severe neutropenia is associated with homozygosity for the UGT1A1*28 or UGT1A1*6 alleles. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 21740478-1 2011 Irinotecan-induced severe neutropenia is associated with homozygosity for the UGT1A1*28 or UGT1A1*6 alleles. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 21740478-2 2011 In this study, we determined the maximum-tolerated dose (MTD) of irinotecan in patients with UGT1A1 polymorphisms. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21740478-14 2011 Patients homozygous for the UGT1A1*28 or UGT1A1*6 allele can receive irinotecan in a starting dose of 150 mg/m(2), but many required dose reductions or delayed treatment in subsequent cycles. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 21740478-14 2011 Patients homozygous for the UGT1A1*28 or UGT1A1*6 allele can receive irinotecan in a starting dose of 150 mg/m(2), but many required dose reductions or delayed treatment in subsequent cycles. Irinotecan 69-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 21524189-5 2011 The potential silybin metabolites were formed in vitro by incubating silybin (i) with the human liver microsomal fraction, (ii) with human hepatocytes and finally (iii) with 12 recombinant UGTs (UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9, 1A10, 2B4, 2B7, 2B15 and 2B17). Silybin 14-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 195-201 21209233-5 2011 Raltegravir is metabolized primarily by UGT1A1 and is not affected by P450 inhibitors or inducers. Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 21209233-6 2011 Inhibitors of UGT1A1 (eg, atazanavir) can increase plasma concentrations of raltegravir, although this increase has not been found to be clinically meaningful. Atazanavir Sulfate 26-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 21209233-6 2011 Inhibitors of UGT1A1 (eg, atazanavir) can increase plasma concentrations of raltegravir, although this increase has not been found to be clinically meaningful. Raltegravir Potassium 76-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 21209233-7 2011 Likewise, inducers of UGT1A1 (eg, rifampin) can reduce plasma concentrations of raltegravir, and the clinical significance of this reduction is being investigated in ongoing clinical studies. Raltegravir Potassium 80-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 21771946-1 2011 BACKGROUND: Gilbert syndrome, a chronic nonhemolytic unconjugated hyperbilirubinemia, is associated with thymine-adenine (TA) insertions in the UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide A1) promoter. thymine-adenine 105-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 21771946-1 2011 BACKGROUND: Gilbert syndrome, a chronic nonhemolytic unconjugated hyperbilirubinemia, is associated with thymine-adenine (TA) insertions in the UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide A1) promoter. thymine-adenine 105-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-204 21771946-1 2011 BACKGROUND: Gilbert syndrome, a chronic nonhemolytic unconjugated hyperbilirubinemia, is associated with thymine-adenine (TA) insertions in the UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide A1) promoter. Tantalum 122-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 21771946-1 2011 BACKGROUND: Gilbert syndrome, a chronic nonhemolytic unconjugated hyperbilirubinemia, is associated with thymine-adenine (TA) insertions in the UGT1A1 (UDP glucuronosyltransferase 1 family, polypeptide A1) promoter. Tantalum 122-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-204 21771946-2 2011 The UGT1A1 promoter genotype also correlates with toxicity induced by the chemotherapeutic drug irinotecan. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 22866151-0 2011 Prevalence of topoisomerase I genetic mutations and UGT1A1 polymorphisms associated with irinotecan in individuals of Asian descent. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 22866151-3 2011 On the other hand, polymorphisms of UGT1A1 have been shown to be associated with CPT-11 toxicity in clinical situations. Irinotecan 81-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 21829131-8 2011 Metabolic clearance of telmisartan in human liver microsomes obtained from individuals with UGT1A1*28/*28 was higher compared with that of UGT1A1*1/*1 (168+-33 vs. 93.3+-27.3 mul/min/mg protein). Telmisartan 23-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 21555506-12 2011 Summing up, we showed that, whereas liver P450 isoenzymes were involved in the metabolism of C-1311 to a limited extent, FMO plays a significant role in the microsomal transformations of this compound, which is also a specific substrate of UGT1A1. C 1311 93-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 240-246 21826053-3 2011 Corydaline also demonstrated moderate inhibition of UGT1A1-mediated 17b-estradiol 3-glucuronidation and UGT1A9-mediated propofol glucuronidation with K(i) values of 57.6 and 37.3 mM, respectively. corydaline 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 21826053-3 2011 Corydaline also demonstrated moderate inhibition of UGT1A1-mediated 17b-estradiol 3-glucuronidation and UGT1A9-mediated propofol glucuronidation with K(i) values of 57.6 and 37.3 mM, respectively. Estradiol 68-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 21624356-0 2011 Simple and precise detection of UGT1A1 polymorphisms with a modified loop-hybrid mobility shift assay using Cy5-labeled loop probes. cyanine dye 5 108-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 21636218-1 2011 Patients with Gilbert syndrome have an impaired function of the enzyme UGT1A1, responsible for the degradation of 4-OH-estrogens. 4-oh 114-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 21585235-6 2011 This achievement has led to personalized irinotecan therapy, reflecting ethnic differences in UGT1A1 genotypes, and possible benefits of genetic testing have also been suggested for gemcitabine and tamoxifen therapy, which still requires further validation. Irinotecan 41-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 21472492-2 2011 We aimed to determine the regioselective glucuronidation of four model flavonols using six expressed human UGT1A isoforms (UGT1A1, 1A3, 1A7, 1A8, 1A9, 1A10). Flavonols 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 21472492-4 2011 RESULTS: UGT1A1 and 1A3 regioselectively metabolized the 7-OH group, whereas UGT1A7, 1A8, 1A9 and 1A10 preferred to glucuronidate the 3-OH group. 7-oh 57-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 21472492-4 2011 RESULTS: UGT1A1 and 1A3 regioselectively metabolized the 7-OH group, whereas UGT1A7, 1A8, 1A9 and 1A10 preferred to glucuronidate the 3-OH group. 3-oh 134-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 21413806-4 2011 UGT 1A1 equally glucuronidated 3-O (glucuronic acid substituted at C-3 hydroxyl group), 7-O, and 4"-O, whereas UGTs 1A8 and 1A9 preferably glucuronidated only 3-O and 7-O positions. 3-o 31-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-7 22169899-1 2011 The UGT1A1 gene encodes a responsible enzyme, UDP-glucuronosyltransferase1A1, for bilirubin metabolism. Bilirubin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 22169899-1 2011 The UGT1A1 gene encodes a responsible enzyme, UDP-glucuronosyltransferase1A1, for bilirubin metabolism. Bilirubin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-76 21287524-2 2011 The UGT1A1 gene encodes for an enzyme that metabolizes irinotecan, and its genetic variants were shown to be associated with increased drug toxicity. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 21287524-12 2011 CONCLUSIONS: The UGT1A1*28 7/7 genotype is strongly associated with severe hematological toxicity and higher hospitalization rate and predicts lower survival of colorectal cancer in users of irinotecan. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 21413806-4 2011 UGT 1A1 equally glucuronidated 3-O (glucuronic acid substituted at C-3 hydroxyl group), 7-O, and 4"-O, whereas UGTs 1A8 and 1A9 preferably glucuronidated only 3-O and 7-O positions. Glucuronic Acid 36-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-7 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21412181-2 2011 Two patients during clinical development met laboratory, but not clinical, criteria for Hy"s law with bilirubin elevations suspected as a result of genetic variation in uridine diphosphoglucose glucuronosyltransferase (UGT1A1) typical of Gilbert syndrome. Bilirubin 102-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 219-225 21412181-7 2011 UGT1A1*28 and three additional single nucleotide polymorphisms showed odds ratios greater than 25 for associations with elevated bilirubin. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21412181-10 2011 Thus, in the absence of other signs of hepatic dysfunction, bilirubin elevations after treatment with tocilizumab have a high probability of association with UGT1A1 polymorphism, which should alleviate concerns of serious hepatotoxicity. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 21507987-9 2011 Regulation of UGT1A1 and UGT1A9 expression showed that the induction of DNA adducts by BaP is directly affected by their expression levels. Benzo(a)pyrene 87-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 29-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21654688-2 2011 Irinotecan is converted into 7-ethyl-10-hydroxycamptothecin (SN-38) by a carboxylsterase and metabolised through uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 21466813-12 2011 Resveratrol at its IC(50) mediated a four-fold induction of UGT1A1 mRNA in a concentration independent manner. Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21317830-0 2011 Association of the UGT1A1-53(TA)n polymorphism with L-thyroxine doses required for thyrotropin suppression in patients with differentiated thyroid cancer. Thyroxine 52-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 21235620-8 2011 Dasatinib and imatinib also inhibited UGT1A1-mediated paracetamol glucuronidation. Dasatinib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 21235620-8 2011 Dasatinib and imatinib also inhibited UGT1A1-mediated paracetamol glucuronidation. Imatinib Mesylate 14-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 21235620-8 2011 Dasatinib and imatinib also inhibited UGT1A1-mediated paracetamol glucuronidation. Acetaminophen 54-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 21317540-4 2011 When UGT1A-expressing HEK293 cells and LS180 cells were treated with curcumin or calphostin C, the exogenous and endogenous UGT1A protein levels in homogenates prepared with Tris-buffered saline were significantly decreased. Curcumin 69-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-10 21317540-4 2011 When UGT1A-expressing HEK293 cells and LS180 cells were treated with curcumin or calphostin C, the exogenous and endogenous UGT1A protein levels in homogenates prepared with Tris-buffered saline were significantly decreased. Curcumin 69-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-129 21317540-4 2011 When UGT1A-expressing HEK293 cells and LS180 cells were treated with curcumin or calphostin C, the exogenous and endogenous UGT1A protein levels in homogenates prepared with Tris-buffered saline were significantly decreased. calphostin C 81-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-10 21317540-4 2011 When UGT1A-expressing HEK293 cells and LS180 cells were treated with curcumin or calphostin C, the exogenous and endogenous UGT1A protein levels in homogenates prepared with Tris-buffered saline were significantly decreased. tris-buffered saline 174-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-129 20852006-2 2011 A prior investigation of raltegravir coadministered with rifampin demonstrated a decrease in plasma concentrations of raltegravir likely secondary to induction of UGT1A1, the enzyme primarily responsible for the metabolism of raltegravir. Raltegravir Potassium 25-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 20852006-2 2011 A prior investigation of raltegravir coadministered with rifampin demonstrated a decrease in plasma concentrations of raltegravir likely secondary to induction of UGT1A1, the enzyme primarily responsible for the metabolism of raltegravir. Rifampin 57-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 20852006-2 2011 A prior investigation of raltegravir coadministered with rifampin demonstrated a decrease in plasma concentrations of raltegravir likely secondary to induction of UGT1A1, the enzyme primarily responsible for the metabolism of raltegravir. Raltegravir Potassium 118-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 20852006-2 2011 A prior investigation of raltegravir coadministered with rifampin demonstrated a decrease in plasma concentrations of raltegravir likely secondary to induction of UGT1A1, the enzyme primarily responsible for the metabolism of raltegravir. Raltegravir Potassium 118-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 20852006-3 2011 Little is known regarding the induction of UGT1A1 by rifabutin, an alternate rifamycin. Rifabutin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 20852006-5 2011 In vitro studies indicate that rifabutin is a less potent inducer of UGT1A1 messenger RNA expression than is rifampin. Rifabutin 31-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 21466813-0 2011 Resveratrol in combination with other dietary polyphenols concomitantly enhances antiproliferation and UGT1A1 induction in Caco-2 cells. Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 21466813-0 2011 Resveratrol in combination with other dietary polyphenols concomitantly enhances antiproliferation and UGT1A1 induction in Caco-2 cells. Polyphenols 46-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 21317830-4 2011 These results are consistent with reduced T4 glucuronidation in patients with low-expression (TA)7 and (TA)8 alleles and provide the first evidence for association between UGT1A1-53(TA)n and T4-dose requirement for thyroid-stimulating hormone suppression in a natural clinical setting. Thyrotropin 215-242 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-178 21245288-5 2011 Higher NFA concentrations (50-100 muM) inhibited UGT1A1 and UGT2B15 but had little effect on the activities of UGT1A3, UGT1A4, UGT1A6, UGT2B4, UGT2B7, and UGT2B17. Niflumic Acid 7-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 21417257-2 2011 The present study investigated the phase II metabolism of feruloylquinic acids with selected human sulfotransferases (SULT1A1 and SULT1E1) and uridine 5"-diphosphoglucuronosyltransferases (UGT1A1 and UGT1A9). 1,3-di-O-feruloylquinic acid 58-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 21266593-2 2011 The development of UGT1A1 and 1A6 was studied in 50 pediatric liver samples using bilirubin, serotonin activity assays, and Western blot as well as pharmacokinetic scaling. Bilirubin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-33 21266593-2 2011 The development of UGT1A1 and 1A6 was studied in 50 pediatric liver samples using bilirubin, serotonin activity assays, and Western blot as well as pharmacokinetic scaling. Serotonin 93-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-33 21538282-5 2011 Linkage and case-control studies of candidate genes and recent genome-wide studies have identified multiple lithogenic genes, in particular the hepatocanalicular cholesterol transporter ABCG5/G8 and the bilirubin conjugating enzyme UGT1A1, as major genetic determinants of gallstones in humans. Bilirubin 203-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 232-238 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-94 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Bilirubin 200-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Bilirubin 200-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-94 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Cholesterol 248-259 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 21353662-7 2011 Variants of the gene encoding UGT1A1 (uridine 5"-diphosphate (UDP)-glucuronosyltransferase 1A1) responsible for bilirubin conjugation were recently associated with risk of gallstones as well as stone bilirubin content, suggesting common factors in cholesterol and pigment gallstone pathogenesis. Cholesterol 248-259 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-94 21368751-3 2011 We have shown in vitro that ezetimibe and tacrolimus may interact in competition for intestinal UGT1A1 and ABCB1 at concentrations reached in gut lumen after oral administration. Ezetimibe 28-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21368751-3 2011 We have shown in vitro that ezetimibe and tacrolimus may interact in competition for intestinal UGT1A1 and ABCB1 at concentrations reached in gut lumen after oral administration. Tacrolimus 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21346002-3 2011 T-5224 was converted to its acyl O-glucuronide (G2) by UGT1A1 and UGT1A3 and to its hydroxyl O-glucuronide (G3) by several UGTs, but it was not metabolized by the P450s. 3-(5-(4-(cyclopentyloxy)-2-hydroxybenzoyl)-2-((3-hydroxy-1,2-benzisoxazol-6-yl)methoxy)phenyl)propionic acid 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 21346002-6 2011 A high correlation was observed between G2 formation activity and UGT1A1-specific activity (beta-estradiol 3-glucuronidation) in seven individual HLM. Estradiol 92-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 21513526-2 2011 Previous genetic studies have focused on the UGT1A1 gene, associating common variation in the coding or promoter regions with qualitative assessments of bilirubin (i.e. significantly elevated or not). Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 21513526-5 2011 We apply this approach to the UGT1A1 gene--exploring the contribution of both rare and common variants to early bilirubin changes. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 21513526-9 2011 CONCLUSIONS: Collectively, rare UGT1A1 coding variants do not appear to play a prominent role in determining early bilirubin levels; however common variants in the 3" UTR of UGT1A1 may modulate the early bilirubin rise. Bilirubin 204-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 21189330-9 2011 We conclude that limited in vivo metabolism of AR-67 by UGT1A1 may partly explain the absence of AR-67 glucuronides in plasma and hypothesize that UGT1A8- and CYP3A-mediated biotransformation within the gastrointestinal epithelium may provide protective mechanisms against AR-67 gastrointestinal toxicity. Argon 47-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 21245288-9 2011 Consistent with previous reports, APAP was glucuronidated by recombinant UGT1A1, UGT1A6, UGT1A9, and UGT2B15. Acetaminophen 34-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 21245288-10 2011 NFA concentrations in the range of 2.5 to 100 muM inhibited APAP glucuronidation by UGT1A1, UGT1A9, and UGT2B15 but not by UGT1A6. Niflumic Acid 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 21216137-2 2011 In vitro studies have shown that omeprazole can alter the function of metabolic enzymes and transporters that are involved in the metabolism of irinotecan, such as uridine diphosphate glucuronosyltransferase subfamily 1A1 (UGT1A1), cytochrome P-450 enzymes subfamily 3A (CYP3A) and ATP-binding cassette drug-transporter G2 (ABCG2). Omeprazole 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 223-229 21255081-6 2011 Bilirubin concentrations were significantly higher among individuals with compound heterozygous variations/homozygous variation in the UGT1A1 gene than in those possessing the wild type and heterozygous variation (P < 0.001 for both alpha- and beta-thalassemia heterozygotes). Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 21255081-8 2011 CONCLUSIONS: The difference in bilirubin concentrations between alpha- and beta-thalassemia heterozygotes may be attributable to more bilirubin being produced in beta-thalassemia heterozygotes than in alpha-thalassemia heterozygotes, while variation status of the UGT1A1 gene affects bilirubin concentrations in both alpha- and beta-thalassemia heterozygotes. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 266-272 21216137-2 2011 In vitro studies have shown that omeprazole can alter the function of metabolic enzymes and transporters that are involved in the metabolism of irinotecan, such as uridine diphosphate glucuronosyltransferase subfamily 1A1 (UGT1A1), cytochrome P-450 enzymes subfamily 3A (CYP3A) and ATP-binding cassette drug-transporter G2 (ABCG2). Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 223-229 21255081-8 2011 CONCLUSIONS: The difference in bilirubin concentrations between alpha- and beta-thalassemia heterozygotes may be attributable to more bilirubin being produced in beta-thalassemia heterozygotes than in alpha-thalassemia heterozygotes, while variation status of the UGT1A1 gene affects bilirubin concentrations in both alpha- and beta-thalassemia heterozygotes. Bilirubin 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 266-272 21255081-8 2011 CONCLUSIONS: The difference in bilirubin concentrations between alpha- and beta-thalassemia heterozygotes may be attributable to more bilirubin being produced in beta-thalassemia heterozygotes than in alpha-thalassemia heterozygotes, while variation status of the UGT1A1 gene affects bilirubin concentrations in both alpha- and beta-thalassemia heterozygotes. Bilirubin 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 266-272 21411679-0 2011 Serum bilirubin links UGT1A1*28 polymorphism and predicts long-term cardiovascular events and mortality in chronic hemodialysis patients. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 21366639-4 2011 SN-38 is subsequently detoxified by uridine diphosphate-glycosyltransferase 1, encoded by the UGT1A1 gene. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 22214020-0 2011 [Treatment with CPT-11 based on the UGT1A1 genetic polymorphism]. Irinotecan 16-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 21309756-1 2011 Variation of a short (TA)(n) repeat sequence (rs8175347) covering the TATA box of UGT1A1 (UDP-glucuronosyltransferase1A1) is associated with hyperbilirubinaemia (Gilbert"s syndrome) and adverse drug reactions, and is used for dosage advice for irinotecan. Irinotecan 244-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 21309756-1 2011 Variation of a short (TA)(n) repeat sequence (rs8175347) covering the TATA box of UGT1A1 (UDP-glucuronosyltransferase1A1) is associated with hyperbilirubinaemia (Gilbert"s syndrome) and adverse drug reactions, and is used for dosage advice for irinotecan. Irinotecan 244-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-120 21242184-3 2011 Although she achieved long-term survival of more than 2 years, the last-line chemotherapy, consisting of irinotecan, induced grade 4 febrile neutropenia and her performance status deteriorated even with a reduced dose according to the analysis of UDP-glucuronosyltransferase 1A1. Irinotecan 105-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 247-278 21411679-2 2011 Bilirubin degradation is mainly determined by the activity of hepatic bilirubin uridine diphosphate-glucuronosyltransferase (UGT1A1), which is significantly influenced by a TA-repeat polymorphism in the gene"s promoter, an allele designated UGT1A1*28. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 21411679-2 2011 Bilirubin degradation is mainly determined by the activity of hepatic bilirubin uridine diphosphate-glucuronosyltransferase (UGT1A1), which is significantly influenced by a TA-repeat polymorphism in the gene"s promoter, an allele designated UGT1A1*28. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 241-247 21411679-3 2011 The study aimed to clarify the association between serum bilirubin and UGT1A1*28 polymorphism and their respective effect on outcomes of chronic hemodialysis patients. Bilirubin 57-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 21411679-7 2011 Individuals homozygous for UGT1A1*28 (genotype 7/7) had significantly higher bilirubin levels than those with 6/6 and 7/6 genotypes. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 21411679-10 2011 Moreover, the UGT1A1*28 polymorphism had strong effects on bilirubin levels and the 7/7 genotype might have an important effect on reducing CVEs and death. Bilirubin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 21980965-3 2011 Her HIV treatment was switched to raltegravir which is metabolized by UGT1A1 which does not affect tacrolimus. Raltegravir Potassium 34-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 21123165-4 2011 Glucuronidation of MPA, as well as the expression and probe substrate activity of UGTs primarily responsible for MPA phenol glucuronide (MPAG) formation (UGT1A1 and UGT1A9), and MPA acyl glucuronide (AcMPAG) formation (UGT2B7), was characterized. mycophenolic acid glucuronide 137-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 20177420-2 2011 Irinotecan"s active metabolite is inactivated by UDP-glucuronosyltransferase 1A1 (UGT1A1), which is deficient in Gilbert"s syndrome. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-80 20177420-2 2011 Irinotecan"s active metabolite is inactivated by UDP-glucuronosyltransferase 1A1 (UGT1A1), which is deficient in Gilbert"s syndrome. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 21297369-4 2011 Irinotecan, now used for treatments of many cancers, is metabolically activated to SN-38 and then inactivated to SN-38 glucuronide by a UDP-glucuronosyltransferase UGT1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 21297369-4 2011 Irinotecan, now used for treatments of many cancers, is metabolically activated to SN-38 and then inactivated to SN-38 glucuronide by a UDP-glucuronosyltransferase UGT1A1. 7-ethyl-10-hydroxycamptothecin glucuronide 113-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 21080475-3 2011 Genetic variation within the promoter region of uridine diphosphate glucuronosyltransferase (UGT1A1) on chromosome 2q has been associated with elevated serum bilirubin levels in European populations. Bilirubin 158-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 21080475-10 2011 CONCLUSIONS: Replication of a serum bilirubin QTL on chromosome 2q in American Indians implicates UGT1A1 but further genotyping is warranted to identify additional causative polymorphisms. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 23461146-1 2011 The UDP-glucuronosyltransferase 1A1 gene that encode the enzyme UGT1A1 responsible for glucuronidation undergoes several variations that may affect the enzymatic activity or expression and which are the cause of metabolic disorders related to the glucuronidation of bilirubin, such as Gilbert"s syndrome and Crigler Najjar"s syndrome. Bilirubin 266-275 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-35 23461146-1 2011 The UDP-glucuronosyltransferase 1A1 gene that encode the enzyme UGT1A1 responsible for glucuronidation undergoes several variations that may affect the enzymatic activity or expression and which are the cause of metabolic disorders related to the glucuronidation of bilirubin, such as Gilbert"s syndrome and Crigler Najjar"s syndrome. Bilirubin 266-275 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 22135857-2 2011 The causative mutation in Caucasians is almost exclusively a (TA) dinucleotide insertion in the UGT1A1 promoter. (ta) dinucleotide 61-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 22135857-10 2011 CONCLUSIONS: We developed a new DHPLC assay which is suitable for accurate, automated, highthroughput, robust genotyping of all UGT1A1(TA)n polymorphism variants, compared to a labour intensive and time-consuming SSCP assay. dhplc 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 20680278-8 2011 CONCLUSIONS: Additional in vitro experiments suggest that these results can be explained by induction of UGT1A1 by methimazole, leading to higher SN-38G concentrations. Methimazole 115-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 20680278-8 2011 CONCLUSIONS: Additional in vitro experiments suggest that these results can be explained by induction of UGT1A1 by methimazole, leading to higher SN-38G concentrations. Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 20814789-0 2011 The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. nilotinib 45-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 20814789-0 2011 The small-molecule tyrosine kinase inhibitor nilotinib is a potent noncompetitive inhibitor of the SN-38 glucuronidation by human UGT1A1. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 20814789-1 2011 PURPOSE: Inhibition of the UDP-glucuronosyltransferase (UGT) 1A1 by nilotinib was examined in vitro with SN-38 as a substrate, to estimate the possibility of drug-drug interaction of nilotinib with other medicines predominantly detoxified by UGT1A1. nilotinib 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-64 20814789-1 2011 PURPOSE: Inhibition of the UDP-glucuronosyltransferase (UGT) 1A1 by nilotinib was examined in vitro with SN-38 as a substrate, to estimate the possibility of drug-drug interaction of nilotinib with other medicines predominantly detoxified by UGT1A1. nilotinib 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 242-248 20814789-1 2011 PURPOSE: Inhibition of the UDP-glucuronosyltransferase (UGT) 1A1 by nilotinib was examined in vitro with SN-38 as a substrate, to estimate the possibility of drug-drug interaction of nilotinib with other medicines predominantly detoxified by UGT1A1. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-64 20814789-2 2011 METHODS: Inhibition of UGT1A1-catalyzed SN-38 glucuronidation by nilotinib was examined with human liver microsomes (HLM) and recombinant human UGT1A1 as enzyme sources. Irinotecan 40-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 20814789-2 2011 METHODS: Inhibition of UGT1A1-catalyzed SN-38 glucuronidation by nilotinib was examined with human liver microsomes (HLM) and recombinant human UGT1A1 as enzyme sources. nilotinib 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 20814789-2 2011 METHODS: Inhibition of UGT1A1-catalyzed SN-38 glucuronidation by nilotinib was examined with human liver microsomes (HLM) and recombinant human UGT1A1 as enzyme sources. nilotinib 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. nilotinib 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. nilotinib 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 20814789-4 2011 RESULTS: Nilotinib potently inhibited the SN-38 glucuronidation by human liver microsomal UGT1A1 and recombinant UGT1A1 in a noncompetitive manner, with K(i) values of 0.286 +- 0.0094 and 0.079 +- 0.0029 muM, respectively. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 20814789-5 2011 If a drug that serves as a substrate of UGT1A1 is administered with nilotinib, the area under the plasma concentration-time curve of a drug estimated by using these K(i) values would be two times or higher than that without nilotinib, suggesting drug-drug interactions involving UGT1A1. nilotinib 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 20814789-5 2011 If a drug that serves as a substrate of UGT1A1 is administered with nilotinib, the area under the plasma concentration-time curve of a drug estimated by using these K(i) values would be two times or higher than that without nilotinib, suggesting drug-drug interactions involving UGT1A1. nilotinib 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 279-285 20814789-5 2011 If a drug that serves as a substrate of UGT1A1 is administered with nilotinib, the area under the plasma concentration-time curve of a drug estimated by using these K(i) values would be two times or higher than that without nilotinib, suggesting drug-drug interactions involving UGT1A1. nilotinib 224-233 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 20814789-7 2011 CONCLUSION: We found that nilotinib is a potent noncompetitive inhibitor of human UGT1A1 activity. nilotinib 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 21673874-11 2011 The hyperbilirubinemia seen in both groups may be at least partly related to inhibition of UGT1A1 by sorafenib. Sorafenib 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 21468552-4 2011 UDP-glucuronosyltransferase (UGT)1A1 is a conjugating biotransformation enzyme that plays a role in maintaining the levels of endogenous compounds (e.g., bilirubin) and in handling exogenous compounds, including carcinogens. Bilirubin 154-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-36 21468552-5 2011 It has been demonstrated that the UGT1A1*28 polymorphism plays a predictive role in patients administered an irinotecan-containing regimen. Irinotecan 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 21617725-9 2011 Sequencing analysis of the UGT1A genes found that the patient had variant alleles of UGT1A1*28, UGT1A1*60 and UGT1A9*22, which may lead to decreased glucuronidation and excretion of SN-38, and may account for increased irinotecan-related toxicity. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 21030469-0 2011 Correlation between bilirubin glucuronidation and estradiol-3-gluronidation in the presence of model UDP-glucuronosyltransferase 1A1 substrates/inhibitors. Bilirubin 20-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-132 21030469-1 2011 Inhibition of UDP-glucuronosyltransferase (UGT) 1A1-catalyzed bilirubin glucuronidation by drug compounds may potentially be of clinical concern. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-51 21030469-5 2011 The glucuronidation kinetics of bilirubin and estradiol were carefully characterized with recombinant UGT1A1 expressed in human embryonic kidney 293 cells. Bilirubin 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Irinotecan 13-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21030469-11 2011 In addition, 7-ethyl-10-hydroxycamptothecin, a substrate of UGT1A1 (reported K(m) = 24 muM) seemed to be a weak inhibitor of bilirubin glucuronidation (IC(50) = 356.4 muM) but a partial inhibitor of estradiol-3-glucuronidation. Estradiol 199-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 21614935-1 2011 Bilirubin is glucoronized by uridine diphosphate-glucuronosyl transferase 1A1 (UGT1A1) mainly in the liver, and excreted into bile. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-77 21614935-1 2011 Bilirubin is glucoronized by uridine diphosphate-glucuronosyl transferase 1A1 (UGT1A1) mainly in the liver, and excreted into bile. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 20975617-7 2011 Neonates with nucleotide 211 GA or AA variation in UGT1A1 genotypes had higher peak serum bilirubin levels and higher incidence of hyperbilirubinemia than WTs (GG). Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 20854796-2 2011 A microarrays analysis between sensitive and methotrexate resistant MCF7 and MDA-MB-468 breast cancer cells pointed out the UDP-glucuronosyltransferase 1A (UGT1A) family as a common deregulated node in both cell lines. Methotrexate 45-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-154 20854796-2 2011 A microarrays analysis between sensitive and methotrexate resistant MCF7 and MDA-MB-468 breast cancer cells pointed out the UDP-glucuronosyltransferase 1A (UGT1A) family as a common deregulated node in both cell lines. Methotrexate 45-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-161 22110515-0 2011 Is Ceftriaxone-Induced Biliary Pseudolithiasis Influenced by UDP-Glucuronosyltransferase 1A1 Gene Polymorphisms? Ceftriaxone 3-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-92 22110515-3 2011 We describe 3 children with ceftriaxone-induced pseudolithiasis, who were also carriers of the A(TA)(7)TAA polymorphism of the UGT1A1 gene, implying that a cause and effect relation may exist. Ceftriaxone 28-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 21422672-11 2011 Moreover, octyl gallate, a marker substrate of UGT1A1, competitively inhibited FA glucuronidation mediated by this isoform. octyl gallate 10-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 20926993-1 2010 Raltegravir (RAL) is primarily metabolized by uridine diphosphate-glucorunosyl transferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-94 20865252-2 2011 Ezetimibe is readily absorbed, and undergoes rapid and almost complete glucuronidation by UGT, particularly UGT1A1, in enterocytes during its first pass. Ezetimibe 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 20528568-6 2011 RESULTS: The UGT1A1*28 polymorphism was associated with total cholesterol (T-chol) (p = 0.030; corrected p = 0.060) and low-density lipoprotein cholesterol (LDL-C) levels (p = 0.011, corrected p = 0.022) in premenopausal women. Cholesterol 62-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 20528568-6 2011 RESULTS: The UGT1A1*28 polymorphism was associated with total cholesterol (T-chol) (p = 0.030; corrected p = 0.060) and low-density lipoprotein cholesterol (LDL-C) levels (p = 0.011, corrected p = 0.022) in premenopausal women. t-chol 75-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 20528568-8 2011 CONCLUSION: The data showed the presence of an association between the UGT1A1*28/*28 and SULT1A1*2/*2 and T-chol and LDL-C levels in women with different hormonal status. chol 108-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 20648548-0 2011 Selective regulation of UGT1A1 and SREBP-1c mRNA expression by docosahexaenoic, eicosapentaenoic, and arachidonic acids. docosahexaenoic 63-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 20648548-0 2011 Selective regulation of UGT1A1 and SREBP-1c mRNA expression by docosahexaenoic, eicosapentaenoic, and arachidonic acids. eicosapentaenoic 80-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 20648548-0 2011 Selective regulation of UGT1A1 and SREBP-1c mRNA expression by docosahexaenoic, eicosapentaenoic, and arachidonic acids. Arachidonic Acids 102-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 20648548-8 2011 Nevertheless, when these PUFAs were combined with Vit.E, which by itself did not produce any effect, the result was a reduction of UGT1A1 mRNA with DHA, an increase reverting to basal level with EPA and no variation with AA. dehydroacetic acid 148-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 21351260-1 2010 UDP-glucuronosyltransferase 1A1 (UGT1A1) is an enzyme which catalyses not only the glucuronidation of tobacco smoke carcinogens like benzopyrene, but also of the endogenous substrate bilirubin. Benzopyrenes 133-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 21351260-1 2010 UDP-glucuronosyltransferase 1A1 (UGT1A1) is an enzyme which catalyses not only the glucuronidation of tobacco smoke carcinogens like benzopyrene, but also of the endogenous substrate bilirubin. Bilirubin 183-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 21351260-8 2010 In conclusion, the predicted high activity UGT1A1*1 polymorphism, which results in lower serum levels of the endogenous antioxidant bilirubin, was associated with an increased risk of head and neck cancer. Bilirubin 132-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 21191764-5 2010 Jaceosidin glucuronidation was catalyzed by UGT1A1, UGT1A3, UGT1A7, UGT1A8, UGT1A9, and UGT1A10. jaceosidin glucuronidation 0-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 21191764-6 2010 These results suggest that the pharmacokinetics of jaceosidin may be dramatically affected by polymorphic CYP1A2, UGT1A1, and UGT1A7 responsible for the metabolism of jaceosidin or by the coadministration of relevant CYP1A2 or UGT inhibitors or inducers. jaceosidin 51-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 21191764-6 2010 These results suggest that the pharmacokinetics of jaceosidin may be dramatically affected by polymorphic CYP1A2, UGT1A1, and UGT1A7 responsible for the metabolism of jaceosidin or by the coadministration of relevant CYP1A2 or UGT inhibitors or inducers. jaceosidin 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 21150467-13 2011 The UGT1A1*6 and UGT1A1*27 genotypes might be useful predictors of grade 4 neutropenia in patients who receive irinotecan-based chemotherapy. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 21150467-13 2011 The UGT1A1*6 and UGT1A1*27 genotypes might be useful predictors of grade 4 neutropenia in patients who receive irinotecan-based chemotherapy. Irinotecan 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 21175442-4 2010 Finally, a panel of human liver microsomes (genotyped for UGT1A1*28 allele status) was used to determine the importance of UGT1A1 in the direct glucuronidation of repaglinide. repaglinide 163-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 21175442-10 2010 UGT1A1 plays a significant role in the glucuronidation of repaglinide. repaglinide 58-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 62-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Glucuronides 82-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. repaglinide 176-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 21175442-12 2010 CONCLUSIONS: Inhibition of UGT enzymes, especially UGT1A1, by gemfibrozil and its glucuronide is an additional mechanism to consider when rationalizing the interaction between repaglinide and gemfibrozil. Gemfibrozil 192-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 20837016-5 2010 RESULTS: By using the presence of bilirubin as a phenotype, variants rs6742078 (UGT1A1; P = .003), rs4149056 (SLCO1B1; P = .003), and rs4149000 (SLCO1A2; P = .015) were associated with gallstone composition. Bilirubin 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 20837016-10 2010 CONCLUSIONS: The UGT1A1 Gilbert syndrome variant rs6742078 is associated with gallstone disease in men; further studies are required regarding the sex-specific physiology of bilirubin and bile acid metabolism. Bilirubin 174-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 20837016-10 2010 CONCLUSIONS: The UGT1A1 Gilbert syndrome variant rs6742078 is associated with gallstone disease in men; further studies are required regarding the sex-specific physiology of bilirubin and bile acid metabolism. Bile Acids and Salts 188-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 21284263-1 2010 The aim of the present study was to investigate the inhibitory effects of medroxyprogesterone acetate (MPA) on four important UGT isoforms (UGT1A1, 1A6, 1A9 and 2B7). Medroxyprogesterone Acetate 74-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 21284263-1 2010 The aim of the present study was to investigate the inhibitory effects of medroxyprogesterone acetate (MPA) on four important UGT isoforms (UGT1A1, 1A6, 1A9 and 2B7). Medroxyprogesterone Acetate 103-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 20926993-1 2010 Raltegravir (RAL) is primarily metabolized by uridine diphosphate-glucorunosyl transferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 20926993-2 2010 Atazanavir (ATV), a strong inhibitor of UGT1A1, has been shown to increase plasma concentrations of RAL by approximately 50% in healthy volunteers. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 20926993-2 2010 Atazanavir (ATV), a strong inhibitor of UGT1A1, has been shown to increase plasma concentrations of RAL by approximately 50% in healthy volunteers. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 20926993-8 2010 Notably, patients with ATV AUC 0-12 above the mean or with concentrations exceeding the half maximal inhibitory concentration for UGT1A1 had twofold higher RAL AUCs compared with patients with lower ATV exposure. Atazanavir Sulfate 23-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 20926993-8 2010 Notably, patients with ATV AUC 0-12 above the mean or with concentrations exceeding the half maximal inhibitory concentration for UGT1A1 had twofold higher RAL AUCs compared with patients with lower ATV exposure. Atazanavir Sulfate 199-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 20823282-1 2010 Atazanavir inhibits UDP-glucuronyl-transferase-1A1 (UGT1A1), which metabolizes raltegravir, but the magnitude of steady-state inhibition and role of the UGT1A1 genotype are unknown. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-50 20823282-1 2010 Atazanavir inhibits UDP-glucuronyl-transferase-1A1 (UGT1A1), which metabolizes raltegravir, but the magnitude of steady-state inhibition and role of the UGT1A1 genotype are unknown. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 20823282-1 2010 Atazanavir inhibits UDP-glucuronyl-transferase-1A1 (UGT1A1), which metabolizes raltegravir, but the magnitude of steady-state inhibition and role of the UGT1A1 genotype are unknown. Raltegravir Potassium 79-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-50 20823282-1 2010 Atazanavir inhibits UDP-glucuronyl-transferase-1A1 (UGT1A1), which metabolizes raltegravir, but the magnitude of steady-state inhibition and role of the UGT1A1 genotype are unknown. Raltegravir Potassium 79-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 20668247-0 2010 Bilirubin glucuronidation revisited: proper assay conditions to estimate enzyme kinetics with recombinant UGT1A1. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 20709051-0 2010 The effect of UGT1A1 promoter polymorphism on bilirubin response to hydroxyurea therapy in hemoglobinopathies. Bilirubin 46-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 20709051-0 2010 The effect of UGT1A1 promoter polymorphism on bilirubin response to hydroxyurea therapy in hemoglobinopathies. Hydroxyurea 68-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 20709051-2 2010 However, the effect of hydroxyurea on hyperbilirubinemia in relation to the UGT1A1 gene promoter polymorphism is not known in Indian patients with different hemoglobinopathies. Hydroxyurea 23-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 20668247-1 2010 Bilirubin, an end product of heme catabolism, is primarily eliminated via glucuronic acid conjugation by UGT1A1. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 20668247-1 2010 Bilirubin, an end product of heme catabolism, is primarily eliminated via glucuronic acid conjugation by UGT1A1. Glucuronic Acid 74-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 20668247-2 2010 Impaired bilirubin conjugation, caused by inhibition of UGT1A1, can result in clinical consequences, including jaundice and kernicterus. Bilirubin 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 20668247-3 2010 Thus, evaluation of the ability of new drug candidates to inhibit UGT1A1-catalyzed bilirubin glucuronidation in vitro has become common practice. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 20668247-10 2010 In conclusion, establishment of appropriate incubation conditions (i.e., very low protein concentrations and short incubation times) is necessary to properly characterize the kinetics of bilirubin glucuronidation in a recombinant UGT1A1 system. Bilirubin 187-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 21072184-7 2010 Polymorphisms in ABCC2, ABCG2, UGT1A1, UGT1A9, and SLCO1B1 were found to significantly correlate with flavopiridol PK in univariate analysis. alvocidib 102-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 20890421-11 2010 Our data suggest that atazanavir-associated hyperbilirubinemia is common but transient in a population with low UGT1A1*28 allele frequency. Atazanavir Sulfate 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 20693308-9 2010 SUMMARY: Serum bilirubin has a protective effect on CVD and CVD-related diseases, and UGT1A1 is the major gene controlling serum bilirubin concentrations. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 20890421-0 2010 Incidence of atazanavir-associated hyperbilirubinemia in Korean HIV patients: 30 months follow-up results in a population with low UDP-glucuronosyltransferase1A1*28 allele frequency. Atazanavir Sulfate 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-161 20693308-7 2010 A conditional linkage study indicates that UGT1A1 is the major gene controlling serum bilirubin concentrations, and this finding has been confirmed in recent genomewide association studies. Bilirubin 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 20666626-3 2010 Estradiol-3-O-glucuronide (E-3-G) formation was used as the index of UGT1A1 activity. estradiol-3-o-glucuronide 0-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 20397195-6 2010 UGT1A1, 1A3 and 1A8 had the highest activities and gave rise to the phenolic glucuronide, whereas glucuronidation of the aliphatic hydroxyl group was mostly mediated by UGT2B7 with low activity. Glucuronides 77-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20397195-6 2010 UGT1A1, 1A3 and 1A8 had the highest activities and gave rise to the phenolic glucuronide, whereas glucuronidation of the aliphatic hydroxyl group was mostly mediated by UGT2B7 with low activity. aliphatic hydroxyl 121-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20890421-2 2010 UDP-glucuronosyltransferase 1A1 polymorphism, UGT1A1*28, which is associated with atazanavir-induced hyperbilirubinemia, is less common in Asians than in Caucasians. Atazanavir Sulfate 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 20890421-2 2010 UDP-glucuronosyltransferase 1A1 polymorphism, UGT1A1*28, which is associated with atazanavir-induced hyperbilirubinemia, is less common in Asians than in Caucasians. Atazanavir Sulfate 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 20061399-1 2010 Alterations in the hepatic conjugation of bilirubin due to uridyl-diphosphate-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms have been proposed as risk factors to neonatal jaundice. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-105 20061399-1 2010 Alterations in the hepatic conjugation of bilirubin due to uridyl-diphosphate-glucuronosyltransferase 1A1 (UGT1A1) polymorphisms have been proposed as risk factors to neonatal jaundice. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 20666626-3 2010 Estradiol-3-O-glucuronide (E-3-G) formation was used as the index of UGT1A1 activity. e-3-g 27-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 20639394-4 2010 In a conditional analysis adjusted for the top UGT1A1 variant (rs11891311), another variant in UGT1A1 (rs4148323, P = 1.22 x 10(-121)) remained significant; this suggests that in UGT1A1 at least two independent genetic variations influence the bilirubin levels in the Korean population. Bilirubin 244-253 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 20639394-4 2010 In a conditional analysis adjusted for the top UGT1A1 variant (rs11891311), another variant in UGT1A1 (rs4148323, P = 1.22 x 10(-121)) remained significant; this suggests that in UGT1A1 at least two independent genetic variations influence the bilirubin levels in the Korean population. Bilirubin 244-253 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 20516255-6 2010 The kinetic parameters of BZA-4"-glucuronide formation were determined in liver, duodenum, and jejunum microsomes and with UGT1A1, 1A8, and 1A10, the most active UGT isoforms involved in the glucuronidation of BZA, whereas those of BZA-5-glucuronide were determined with all the enzyme systems except in liver microsomes and in UGT1A1 because the formation of the BZA-5-glucuronide was too low. bza-4"-glucuronide 26-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 20516255-6 2010 The kinetic parameters of BZA-4"-glucuronide formation were determined in liver, duodenum, and jejunum microsomes and with UGT1A1, 1A8, and 1A10, the most active UGT isoforms involved in the glucuronidation of BZA, whereas those of BZA-5-glucuronide were determined with all the enzyme systems except in liver microsomes and in UGT1A1 because the formation of the BZA-5-glucuronide was too low. bza-4"-glucuronide 26-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 328-334 20516255-7 2010 K(m) values in liver, duodenum, and jejunum microsomes and UGT1A1, 1A8, and 1A10, were similar and ranged from 5.1 to 33.1 microM for BZA-4"-glucuronide formation and from 2.5 to 11.1 microM for BZA-5-glucuronide formation. bza-4"-glucuronide 134-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 20643254-0 2010 Relation of ADRB1, CYP2D6, and UGT1A1 polymorphisms with dose of, and response to, carvedilol or metoprolol therapy in patients with chronic heart failure. Carvedilol 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 21092520-3 2010 The uridine diphosphate-glucuronosyl transferase 1A1 (UGT 1A1) gene controls bilirubin conjugation by determining the structure of the enzyme glucuronosyltransferase, which is synthesized in the hepatocyte. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-52 21092520-3 2010 The uridine diphosphate-glucuronosyl transferase 1A1 (UGT 1A1) gene controls bilirubin conjugation by determining the structure of the enzyme glucuronosyltransferase, which is synthesized in the hepatocyte. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-61 20546738-8 2010 RESULTS: In Gunn rats, bilirubin levels normalized 2 weeks after administration of mTTR.hUGT1A1. Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-95 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. 7-hydroxy-4-trifluoromethylcoumarin 86-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. 7-hydroxy-4-trifluoromethylcoumarin 123-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. Estradiol 131-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. e-3oh 164-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. Irinotecan 175-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-5 2010 The enzymatic properties of UGT1A1 proteins were characterized by kinetic analysis of 7-hydroxy-4-trifluoromethylcoumarin (7-HFC), estradiol at 3-hydroxy position (E-3OH) and 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation. Irinotecan 207-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20620155-9 2010 In recombinant UGT1A1 enzymes expressed in insect cells, the kinetics of 7-HFC, E-3OH and SN-38 glucuronidation fitted the substrate inhibition (7-HFC glucuronidation) or Hill equation (E-3OH and SN-38 glucuronidation), and each glucuronidation showed the same kinetic profile between humans and cynomolgus monkeys. 3oh 82-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 20620155-9 2010 In recombinant UGT1A1 enzymes expressed in insect cells, the kinetics of 7-HFC, E-3OH and SN-38 glucuronidation fitted the substrate inhibition (7-HFC glucuronidation) or Hill equation (E-3OH and SN-38 glucuronidation), and each glucuronidation showed the same kinetic profile between humans and cynomolgus monkeys. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 20620155-9 2010 In recombinant UGT1A1 enzymes expressed in insect cells, the kinetics of 7-HFC, E-3OH and SN-38 glucuronidation fitted the substrate inhibition (7-HFC glucuronidation) or Hill equation (E-3OH and SN-38 glucuronidation), and each glucuronidation showed the same kinetic profile between humans and cynomolgus monkeys. Tin 90-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 20643254-0 2010 Relation of ADRB1, CYP2D6, and UGT1A1 polymorphisms with dose of, and response to, carvedilol or metoprolol therapy in patients with chronic heart failure. Metoprolol 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 20643254-2 2010 The purpose of the present study was to determine whether specific genetic polymorphisms in ADRB1 (encoding the beta1-adrenergic receptor), CYP2D6, and UGT1A1 correlated with dose of, or response to, metoprolol or carvedilol treatment in patients with heart failure. Metoprolol 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 20643254-2 2010 The purpose of the present study was to determine whether specific genetic polymorphisms in ADRB1 (encoding the beta1-adrenergic receptor), CYP2D6, and UGT1A1 correlated with dose of, or response to, metoprolol or carvedilol treatment in patients with heart failure. Carvedilol 214-224 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 20412608-9 2010 In addition, UGT1A1 expression induced by UCB in Caco-2 cells could ameliorate the cytotoxicity of UCB. ucb 42-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 20406168-5 2010 In recent years, genetic polymorphisms in UDP-glucuronosyltransferase (UGT) 1A1, an enzyme involved in SN-38 glucuronidation, has been linked to interindividual pharmacokinetic variability and irinotecan toxicity. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-79 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 158-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-89 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 158-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 190-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-89 20653675-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Association of UDP-glucuronosyltransferase 1A1 (UGT1A1) genetic polymorphisms *6 and *28 with reduced clearance of SN-38 and severe neutropenia in irinotecan therapy was demonstrated in Japanese cancer patients. Irinotecan 190-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 20562211-0 2010 Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 20562211-1 2010 PURPOSE: A previous meta-analysis showed that the association between the UGT1A1*28 genotype and irinotecan-induced neutropenia was influenced by irinotecan dose and that the risk of neutropenia was similar at low doses for patients with all genotypes. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 20562211-1 2010 PURPOSE: A previous meta-analysis showed that the association between the UGT1A1*28 genotype and irinotecan-induced neutropenia was influenced by irinotecan dose and that the risk of neutropenia was similar at low doses for patients with all genotypes. Irinotecan 146-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 20562211-5 2010 The association between UGT1A1*28 and the relative extent of glucuronidation (REG) of SN-38 was also examined. Irinotecan 86-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 20562211-11 2010 We found the weighted mean difference of REG (UGT1A1*1/*1 or UGT1A1*1/*28 versus UGT1A1*28/*28) increased with increasing dose of irinotecan. Irinotecan 130-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 20562211-12 2010 CONCLUSIONS: In conclusion, the UGT1A1*28/*28 genotype was associated with an increased risk of neutropenia not only at medium or high doses of irinotecan but also at low doses. Irinotecan 144-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 20562211-13 2010 The dose-dependent manner of SN-38 glucuronidation explained why the association between UGT1A1*28 and neutropenia was dose dependent. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 20406168-5 2010 In recent years, genetic polymorphisms in UDP-glucuronosyltransferase (UGT) 1A1, an enzyme involved in SN-38 glucuronidation, has been linked to interindividual pharmacokinetic variability and irinotecan toxicity. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-79 20063115-10 2010 SN-38 C(max) and AUC in EIAC patients receiving 400 mg/m(2) irinotecan were 20.9 ng/ml and 212 ng/ml h, and in non-EIAC patients receiving 125 mg/m(2), 15.5 ng/ml and 207 ng/ml h. SN-38 AUC varied by UGT1A1*28 status in non-EIAC patients. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 200-206 20562445-2 2010 A major reason for the variability is a common promoter polymorphism, UGT1A1*28, which reduces the transcription of the enzyme that conjugates bilirubin, UDP-glucuronosyltransferase 1A1. Bilirubin 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 20562445-2 2010 A major reason for the variability is a common promoter polymorphism, UGT1A1*28, which reduces the transcription of the enzyme that conjugates bilirubin, UDP-glucuronosyltransferase 1A1. Bilirubin 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-185 20063115-12 2010 Non-EIAC patients with UGT1A1*28 variant alleles appear particularly sensitive to toxicity from irinotecan. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 20580994-0 2010 The UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Erlotinib Hydrochloride 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 20637356-4 2010 The phenotyping of DPD and UGT1A1 activities, and to a lesser extent, its genotyping, appears as the most useful tool in terms of prediction of toxicities induced by two major drugs: 5-FU and irinotecan. Fluorouracil 183-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 20637356-4 2010 The phenotyping of DPD and UGT1A1 activities, and to a lesser extent, its genotyping, appears as the most useful tool in terms of prediction of toxicities induced by two major drugs: 5-FU and irinotecan. Irinotecan 192-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 20335017-9 2010 The risk of severe diarrhoea at medium and high irinotecan doses was higher among patients with a UGT1A1*28/*28 genotype than among those with a UGT1A1*1/*1 genotype (OR=3.69, 95% confidence interval [CI]=2.00-6.83; P<0.001). Irinotecan 48-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 20335017-12 2010 In conclusion, patients carrying UGT1A1*28 allele(s) are at an increased risk of irinotecan-induced severe diarrhoea. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 20207085-5 2010 Recent work has linked PNJ to a specific enzyme polymorphism, a variation of the UGT1A1 gene, in the glucoronidation pathway which is essential for bilirubin metabolism and is strongly correlated with ethnic origin. glucoronidation 101-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 20207085-5 2010 Recent work has linked PNJ to a specific enzyme polymorphism, a variation of the UGT1A1 gene, in the glucoronidation pathway which is essential for bilirubin metabolism and is strongly correlated with ethnic origin. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 20661248-1 2010 BACKGROUND: Accurate description of current practice within advanced colorectal cancer (CRC) specialties were needed to inform an economic evaluation of the UGT1A1 pharmacogenetic test for irinotecan in the United Kingdom. Irinotecan 189-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 20504240-1 2010 BACKGROUND: High prevalence of severe atazanavir-associated hyperbilirubinemia in Asians with low prevalence of the UDP-glucuronosyltransferase (UGT)1A1*28 polymorphism suggests the importance of genetic factors other than UGT1A1*28 for atazanavir-associated hyperbilirubinemia in these populations. Atazanavir Sulfate 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 223-229 20504240-1 2010 BACKGROUND: High prevalence of severe atazanavir-associated hyperbilirubinemia in Asians with low prevalence of the UDP-glucuronosyltransferase (UGT)1A1*28 polymorphism suggests the importance of genetic factors other than UGT1A1*28 for atazanavir-associated hyperbilirubinemia in these populations. Atazanavir Sulfate 237-247 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-152 20504240-10 2010 CONCLUSION: The MDR1 G2677T/A variation and UGT1A1*28 are independent risk factors for severe atazanavir-associated hyperbilirubinemia in Korean human immunodeficiency virus-infected patients. Atazanavir Sulfate 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 20335017-0 2010 Dose-dependent association between UGT1A1*28 polymorphism and irinotecan-induced diarrhoea: a meta-analysis. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 20335017-2 2010 The associations between the UGT1A1*28 polymorphism and irinotecan-induced diarrhoea remains controversial because of conflicting data in the literature. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 20580994-0 2010 The UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Irinotecan 65-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 20580994-6 2010 Our data showed that erlotinib exhibited potent non-competitive inhibition against SN-38 glucuronidation in pooled HLMs and UGT1A1. Erlotinib Hydrochloride 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 20580994-6 2010 Our data showed that erlotinib exhibited potent non-competitive inhibition against SN-38 glucuronidation in pooled HLMs and UGT1A1. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 20580994-7 2010 Using the physiological and pharmacokinetic parameters obtained from the literature, we estimated the in vivo concentrations of unbound erlotinib available for UGT1A1 active site and thus the AUC ratios of SN-38 were also quantitatively predicted. Erlotinib Hydrochloride 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 20580994-9 2010 Significant correlations were observed between SN-38 glucuronidation activity in the presence of erlotinib and UGT1A1*28 in 52 Caucasian liver microsomes. Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 20580994-10 2010 Our results suggest that erlotinib is a potent inhibitor of SN-38 glucuronidation via UGT1A1 inhibition. Erlotinib Hydrochloride 25-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 20580994-10 2010 Our results suggest that erlotinib is a potent inhibitor of SN-38 glucuronidation via UGT1A1 inhibition. Irinotecan 60-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 20580994-12 2010 UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Erlotinib Hydrochloride 39-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20580994-12 2010 UGT1A1*28 polymorphism correlates with erlotinib"s effect on SN-38 glucuronidation. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20435641-3 2010 To elucidate these molecular mechanisms, we simulated and analysed the glucuronidation of wild-type UGT1A1 and six UGT1A1 mutants, with bilirubin as the substrate. Bilirubin 136-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 20308029-2 2010 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is a main metabolic pathway of estrogen detoxification in steroid target tissues, such as the prostate. Steroids 140-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-64 20979879-3 2010 The phenobarbital-responsive enhancer module (PBREM), TATA box and common mutation sites in exons of UGT1A1 gene were amplified by polymerase chain reaction (PCR) and the products screened by direct DNA sequencing. Phenobarbital 4-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 20297805-4 2010 The results also indicated that UGT1A1, UGT1A7, UGT1A8, UGT1A9, UGT1A10 and UGT2B7 are the most important six UGT isoforms for metabolizing the chosen flavones. Flavones 151-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 20602618-3 2010 Although multiple genes may play a role in irinotecan activity, the majority of evidence to date suggests that variation in expression of UGT1A1 caused by a common promoter polymorphism (UGT1A1*28) is strongly associated with toxicity; however, this link is dose dependent. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 20602618-3 2010 Although multiple genes may play a role in irinotecan activity, the majority of evidence to date suggests that variation in expression of UGT1A1 caused by a common promoter polymorphism (UGT1A1*28) is strongly associated with toxicity; however, this link is dose dependent. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 20370320-2 2010 Several clinical and epidemiological studies have been carried out on key enzymes generating and eliminating bilirubin (heme oxygenase-1 and UDP-glucuronosyltransferase UGT1A1, respectively) and their regulation by the AhR. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 20308029-2 2010 Glucuronidation, mediated by the UDP-glucuronosyltransferase 1A1 (UGT1A1) enzyme, is a main metabolic pathway of estrogen detoxification in steroid target tissues, such as the prostate. Steroids 140-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 20215562-4 2010 Here, we present a homology model of the complete monomeric human UGT1A1, the enzyme that catalyzes bilirubin glucuronidation. Bilirubin 100-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 20022574-2 2010 In particular, polymorphisms across three genes involved in bilirubin production and metabolism [glucose-6-phosphate dehydrogenase (G6PD), uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and solute carrier organic anion transporter polypeptide 1B1 (SLCO1B1)] may interact with each other and/or environmental contributors to produce significant hyperbilirubinemia. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-187 20022574-2 2010 In particular, polymorphisms across three genes involved in bilirubin production and metabolism [glucose-6-phosphate dehydrogenase (G6PD), uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and solute carrier organic anion transporter polypeptide 1B1 (SLCO1B1)] may interact with each other and/or environmental contributors to produce significant hyperbilirubinemia. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 20207827-0 2010 Pegvisomant-induced liver injury is related to the UGT1A1*28 polymorphism of Gilbert"s syndrome. pegvisomant 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-57 19771428-2 2010 METHODS: Associations between transporter haplotypes/variations along with UGT1A1*28 or *6 and SN-38 area under the time-concentration curve (AUC) or neutropenia were examined in irinotecan monotherapy (55 patients) and irinotecan-cisplatin-combination therapy (62 patients). Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 20207827-12 2010 CONCLUSIONS: The UGT1A1*28 genotype and male gender predict an increased incidence of LFT abnormalities during PEG therapy in acromegaly. pegvisomant 111-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 20056724-4 2010 UGT1A9, UGT1A1, UGT1A7, UGT1A8, and UGT1A3 are the major enzymes catalyzing hesperetin glucuronidation, the latter only producing 7-O-glucuronide, whereas UGT1A7 produced mainly 3"-O-glucuronide. hesperetin 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 20513938-3 2010 Patients with genetic variations in UGT1A1 and DPYD genes are hypersensitive to Irinotecan and 5-Fluorouracil (5FU) respectively. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 20513938-3 2010 Patients with genetic variations in UGT1A1 and DPYD genes are hypersensitive to Irinotecan and 5-Fluorouracil (5FU) respectively. Fluorouracil 95-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 20513938-3 2010 Patients with genetic variations in UGT1A1 and DPYD genes are hypersensitive to Irinotecan and 5-Fluorouracil (5FU) respectively. Fluorouracil 111-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 20389299-0 2010 Pazopanib-induced hyperbilirubinemia is associated with Gilbert"s syndrome UGT1A1 polymorphism. pazopanib 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 20389299-7 2010 The Gilbert"s uridine-diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1) TA-repeat polymorphism was significantly associated with pazopanib-induced hyperbilirubinemia in the phase II study. pazopanib 137-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-70 20389299-7 2010 The Gilbert"s uridine-diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1) TA-repeat polymorphism was significantly associated with pazopanib-induced hyperbilirubinemia in the phase II study. pazopanib 137-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 20389299-12 2010 CONCLUSIONS: The UGT1A1 polymorphism is frequently associated with pazopanib-induced hyperbilirubinemia. pazopanib 67-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 20056724-4 2010 UGT1A9, UGT1A1, UGT1A7, UGT1A8, and UGT1A3 are the major enzymes catalyzing hesperetin glucuronidation, the latter only producing 7-O-glucuronide, whereas UGT1A7 produced mainly 3"-O-glucuronide. 7-o-glucuronide 130-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 20056724-4 2010 UGT1A9, UGT1A1, UGT1A7, UGT1A8, and UGT1A3 are the major enzymes catalyzing hesperetin glucuronidation, the latter only producing 7-O-glucuronide, whereas UGT1A7 produced mainly 3"-O-glucuronide. 3"-o-glucuronide 178-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 20511137-3 2010 Numerous variants have been found in UGT1A1 , the main conjugating enzyme of bilirubin and drugs such as the anticancer drug irinotecan. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 20511137-3 2010 Numerous variants have been found in UGT1A1 , the main conjugating enzyme of bilirubin and drugs such as the anticancer drug irinotecan. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 20511137-4 2010 Recently, the US Food and Drug Administration (FDA) recommended testing for the presence of UGT1A1*28 , an allele correlated with decreased transcriptional activity, to predict patients at risk of irinotecan toxicity. Irinotecan 197-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 20197307-2 2010 The constitutive androstane receptor (CAR) is an orphan nuclear receptor that has been shown to participate in the activation of the uridine diphosphate-5"-glucuronosyltransferase 1A1 (UGT1A1) gene, which plays an important role in bilirubin clearance. Bilirubin 232-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-183 20179984-0 2010 Indispensability of UGT1A1*6 genotyping in Japanese cancer patients treated with irinotecan. Irinotecan 81-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 20197307-2 2010 The constitutive androstane receptor (CAR) is an orphan nuclear receptor that has been shown to participate in the activation of the uridine diphosphate-5"-glucuronosyltransferase 1A1 (UGT1A1) gene, which plays an important role in bilirubin clearance. Bilirubin 232-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 20197307-7 2010 These data suggest that OSM might play a role in bilirubin metabolism via the CAR-UGT1A1 pathway. Bilirubin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 19932091-1 2010 BACKGROUND: The basis of Gilbert"s syndrome is a 70% reduction in bilirubin glucuronidation which, in the Caucasian population, is the result of a homozygous TA insertion into the promoter region of the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene (UGT1A128 allele). Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 20057336-1 2010 The uridine diphosphoglucuronate-glucuronosyltransferase 1A1 (UGT1A1) gene encodes the enzyme responsible for bilirubin glucuronidation. Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-60 20057336-1 2010 The uridine diphosphoglucuronate-glucuronosyltransferase 1A1 (UGT1A1) gene encodes the enzyme responsible for bilirubin glucuronidation. Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 20194756-1 2010 High levels of unconjugated bilirubin (UCB) in newborn children is associated with a reduction in hepatic UDP glucuronosyltransferase (UGT) 1A1 activity that can lead to CNS toxicity, brain damage, and even death. Bilirubin 28-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-143 20194756-1 2010 High levels of unconjugated bilirubin (UCB) in newborn children is associated with a reduction in hepatic UDP glucuronosyltransferase (UGT) 1A1 activity that can lead to CNS toxicity, brain damage, and even death. ucb 39-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-143 20145913-4 2010 The assays are based on analysis and quantification by high-performance liquid chromatography-tandem mass spectrometry of glucuronides formed from selective probe substrates, namely, beta-estradiol (UGT1A1, 3-glucuronide), 1-naphthol (UGT1A6), propofol (UGT1A9), and naloxone (UGT2B7). Glucuronides 122-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 20145913-4 2010 The assays are based on analysis and quantification by high-performance liquid chromatography-tandem mass spectrometry of glucuronides formed from selective probe substrates, namely, beta-estradiol (UGT1A1, 3-glucuronide), 1-naphthol (UGT1A6), propofol (UGT1A9), and naloxone (UGT2B7). Estradiol 183-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 20235794-2 2010 Irinotecan is one of the models for personalized medicine based on pharmacogenetics, and a number of clinical studies have revealed significant associations between UGT1A1*28 and irinotecan toxicity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 20235794-2 2010 Irinotecan is one of the models for personalized medicine based on pharmacogenetics, and a number of clinical studies have revealed significant associations between UGT1A1*28 and irinotecan toxicity. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 20038727-1 2010 PURPOSE We aimed to identify the maximum-tolerated dose (MTD) of irinotecan in patients with cancer with UGT1A1*1/*1 and *1/*28 genotypes. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 19996319-2 2010 Initially, using an inducible system that mimics the relative abundance of isoforms 1 and 2 of UGT1A1 in human tissues, the rates of formation of glucuronides were significantly reduced. Glucuronides 146-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 19953640-3 2010 Crigler-Najjar syndrome is another rare disorder of bilirubin metabolism caused by mutation in the gene coding the enzyme UGT1A1. Bilirubin 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 19931474-0 2010 Hereditary spherocytosis and the (TA)nTAA polymorphism of UGT1A1 gene promoter region--a comparison of the bilirubin plasmatic levels in the different clinical forms. Bilirubin 107-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 20028383-1 2010 Although individuals carrying the UGT1A1 allele *28 have an increased risk of severe toxicities associated with irinotecan, no phase I study has been conducted based on the polymorphism. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 20028383-12 2010 The recommended starting doses of biweekly irinotecan for phase II/III were 150 mg/m(2) for patients with the UGT1A1 6/6 genotype and 70 mg/m(2) for those with the 6/7 genotype, respectively. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 20235794-5 2010 This article provides an overview of recent progress in irinotecan pharmacogenetics and discusses the clinical significance of the UGT1A1 genotype/haplotype with regard to severe irinotecan toxicity. Irinotecan 179-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 19932091-1 2010 BACKGROUND: The basis of Gilbert"s syndrome is a 70% reduction in bilirubin glucuronidation which, in the Caucasian population, is the result of a homozygous TA insertion into the promoter region of the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene (UGT1A128 allele). Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 236-242 19744523-8 2010 Other well-established associations include CYP2B6 alleles and efavirenz central nervous system side effects, UGT1A1 alleles and atazanavir-associated hyperbilirubinemia and HLA class II allele HLA-DRB*0101 and nevirapine-associated hypersensitivity. Atazanavir Sulfate 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 99-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-43 20096102-1 2010 BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 20096102-3 2010 Interestingly, one of these CpG dinucleotides (CpG -4) is found close to a HNF1 response element (HRE), known to be involved in activation of UGT1A1 gene expression, and within an upstream stimulating factor (USF) binding site. Dinucleoside Phosphates 32-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 20000589-3 2010 Screening of indole metabolites has identified indoxyl 3-sulfate (I3S) as a potent endogenous ligand that selectively activates the human AHR at nanomolar concentrations in primary human hepatocytes, regulating transcription of multiple genes, including CYP1A1, CYP1A2, CYP1B1, UGT1A1, UGT1A6, IL6, and SAA1. indoxyl 3-sulfate 47-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 278-284 20000589-3 2010 Screening of indole metabolites has identified indoxyl 3-sulfate (I3S) as a potent endogenous ligand that selectively activates the human AHR at nanomolar concentrations in primary human hepatocytes, regulating transcription of multiple genes, including CYP1A1, CYP1A2, CYP1B1, UGT1A1, UGT1A6, IL6, and SAA1. inositol 1,3,4-trisphosphate 66-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 278-284 20371435-0 2010 Coexistence of HFE and rare UGT1A1 genes mutations in patients with iron overload related liver injury. Iron 68-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 19788348-9 2010 7-OHWAR was a high-affinity substrate for UGT1A1, compared to other UGTs. 7-ohwar 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 19788348-10 2010 UGT1A1 and UGT1A10 also glucuronidated 6-OHWAR. 6-ohwar 39-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19955297-8 2010 DISCUSSION: Atazanavir is an inhibitor of the bilirubin-conjugating enzyme UGT1A1 and has been frequently linked to the occurrence of hyperbilirubinemia without complications. Atazanavir Sulfate 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 19955297-8 2010 DISCUSSION: Atazanavir is an inhibitor of the bilirubin-conjugating enzyme UGT1A1 and has been frequently linked to the occurrence of hyperbilirubinemia without complications. Bilirubin 46-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 19850672-5 2010 Erlotinib exhibited selective potent competitive inhibition against 4-MU glucuronidation by UGT1A1, and gefitinib demonstrated a wide range of inhibition against UGT-mediated 4-MU glucuronidation, particularly against UGT1A1, UGT1A7, UGT1A9, and UGT2B7. Erlotinib Hydrochloride 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19796502-2 2010 Among these, urea cycle disorders leading to the impairment to detoxify ammonia and Crigler-Najjar Syndrome type I, a deficiency in the hepatic UDP-glucuronosyltransferase 1A1 present the highest incidence. Ammonia 72-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-175 19796502-6 2010 Moreover, the evaluation of urea synthesis from ammonia and UDP-glucuronosyltransferase 1A1 activity, a newly developed assay using beta-estradiol as substrate, allows the possibility of customizing cell preparation for receptors with urea cycle disorders or Crigler-Najjar Syndrome type I. Estradiol 132-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-91 20948202-2 2010 A major cause of elevated plasma bilirubin is the common UGT1A1*28 promoter polymorphism in the gene of the bilirubin-conjugating enzyme UDP-glucuronosyltransferase 1A1, which reduces transcription by 70%. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 20948202-2 2010 A major cause of elevated plasma bilirubin is the common UGT1A1*28 promoter polymorphism in the gene of the bilirubin-conjugating enzyme UDP-glucuronosyltransferase 1A1, which reduces transcription by 70%. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-168 20948202-2 2010 A major cause of elevated plasma bilirubin is the common UGT1A1*28 promoter polymorphism in the gene of the bilirubin-conjugating enzyme UDP-glucuronosyltransferase 1A1, which reduces transcription by 70%. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 20948202-2 2010 A major cause of elevated plasma bilirubin is the common UGT1A1*28 promoter polymorphism in the gene of the bilirubin-conjugating enzyme UDP-glucuronosyltransferase 1A1, which reduces transcription by 70%. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-168 20948202-8 2010 The UGT1A1*28 polymorphism (allele frequency 30%) showed a strong gene-dose relationship with bilirubin levels both among cases and referents, but was not associated with risk for stroke. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19794410-5 2010 Surprisingly, lactone formation was significantly higher in carriers of UGT1A1*28, an allele that is associated with lower UGT1A1 expression. Lactones 14-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 19794410-5 2010 Surprisingly, lactone formation was significantly higher in carriers of UGT1A1*28, an allele that is associated with lower UGT1A1 expression. Lactones 14-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 19797611-2 2010 Treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) (an extracellular signal-regulated kinase inhibitor) suppressed the HGF-induced expression of UGT1A1 and CYP2B6, as well as p16, p21, and p27 in HepG2 cells. u0124 15-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 19797611-2 2010 Treatment with 1,4-diamino-2,3-dicyano-1,4-bis(methylthio)butadiene (U0126) (an extracellular signal-regulated kinase inhibitor) suppressed the HGF-induced expression of UGT1A1 and CYP2B6, as well as p16, p21, and p27 in HepG2 cells. U 0126 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 19797611-3 2010 The CDK inhibitor roscovitine also enhanced the expression of UGT1A1, CYP2B6, and CYP3A4. Roscovitine 18-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 19835560-6 2010 Variant alleles of UGT1A1 are less capable of conjugating and eliminating SN-38, the active form of the topoisomerase inhibitor irinotecan, and defective alleles for NAT2 are responsible for the well-described acetylation polymorphism that leads to impaired clearance of isoniazid and other agents. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 19835560-6 2010 Variant alleles of UGT1A1 are less capable of conjugating and eliminating SN-38, the active form of the topoisomerase inhibitor irinotecan, and defective alleles for NAT2 are responsible for the well-described acetylation polymorphism that leads to impaired clearance of isoniazid and other agents. Irinotecan 128-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 19835560-6 2010 Variant alleles of UGT1A1 are less capable of conjugating and eliminating SN-38, the active form of the topoisomerase inhibitor irinotecan, and defective alleles for NAT2 are responsible for the well-described acetylation polymorphism that leads to impaired clearance of isoniazid and other agents. Isoniazid 271-280 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 19797611-6 2010 In cells arrested in S phase by a thymidine block and then released into a synchronous cell cycle, there was a clear dissociation among the activation of CDK2 and the expression of UGT1A1, CYP2B6, and CYP3A4. Thymidine 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 19850672-5 2010 Erlotinib exhibited selective potent competitive inhibition against 4-MU glucuronidation by UGT1A1, and gefitinib demonstrated a wide range of inhibition against UGT-mediated 4-MU glucuronidation, particularly against UGT1A1, UGT1A7, UGT1A9, and UGT2B7. Erlotinib Hydrochloride 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 218-224 19850672-5 2010 Erlotinib exhibited selective potent competitive inhibition against 4-MU glucuronidation by UGT1A1, and gefitinib demonstrated a wide range of inhibition against UGT-mediated 4-MU glucuronidation, particularly against UGT1A1, UGT1A7, UGT1A9, and UGT2B7. Gefitinib 104-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 218-224 19850672-7 2010 We estimated that coadministration of erlotinib at 100 mg/day or higher doses may result in at least a 30% increase in the AUC of drugs predominantly cleared by UGT1A1. Erlotinib Hydrochloride 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 19850672-8 2010 Thus, the coadministration of erlotinib with drugs primarily cleared by UGT1A1 may result in potential DDI. Erlotinib Hydrochloride 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 20201778-3 2010 The system was then validated using known ligands of PXR, rifampicin (RIF), clotrimazole (CLOT) sulfinpyrazone (SPZ) and phenobarbital (PB), which produced dose dependent induction of UGT1A1 luciferase activity by 4.4, 5.3, 4.7 and 3.7 fold, respectively, relative to the vehicle control, 0.1 % dimethylsulfoxide (DMSO). clotrimazole (clot) sulfinpyrazone 76-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 184-190 19830808-7 2010 In addition to bilirubin, we also determined their activity toward eight other UGT1A1 substrates. Bilirubin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 20201778-3 2010 The system was then validated using known ligands of PXR, rifampicin (RIF), clotrimazole (CLOT) sulfinpyrazone (SPZ) and phenobarbital (PB), which produced dose dependent induction of UGT1A1 luciferase activity by 4.4, 5.3, 4.7 and 3.7 fold, respectively, relative to the vehicle control, 0.1 % dimethylsulfoxide (DMSO). Phenobarbital 121-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 184-190 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. a-naphthoflavone 40-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. a-naphthoflavone 40-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. a-naph 58-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. a-naph 58-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. beta-Naphthoflavone 67-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. beta-Naphthoflavone 67-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. b-naph 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. b-naph 85-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. 3-methylchloranthene 97-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. 3-methylchloranthene 97-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. Methylcholanthrene 119-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 20201778-4 2010 Aryl hydrocarbon receptor (AhR) ligands a-naphthoflavone (a-Naph), b-naphthoflavone (b-Naph) and 3-methylchloranthene (3-MC) increased UGT1A1 luciferase activity in a concentration dependent manner resulting in 17.2, 11.3 and 6.1 fold, respectively, at their highest concentrations, suggesting that endogenous AhR is also involved in the regulation of the UGT1A1 reporter construct in HepG2 cells. Methylcholanthrene 119-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 356-362 20722998-0 2010 Glucuronidation of piceatannol by human liver microsomes: major role of UGT1A1, UGT1A8 and UGT1A10. 3,3',4,5'-tetrahydroxystilbene 19-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 19475557-9 2010 Coexpression of UGT2B7 also affected the kinetics of estradiol 3-O-glucuronidation by UGT1A1, imipramine N-glucuronidation by UGT1A4, serotonin O-glucuronidation by UGT1A6, and propofol O-glucuronidation by UGT1A9. estradiol 3-o 53-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 20402064-4 2010 Thymine-adenine (TA) repeats in the promoter region of the UGT1A1 gene were investigated by polymerase chain reaction (PCR)-based non-radioactive DNA sequencing. thymine-adenine 0-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 21486535-1 2010 PURPOSE: Functional polymorphisms of the UGT1A1 gene, particularly the UGT1A1*28 variant, are associated with the severity of the bone marrow suppression in patients with metastatic colorectal cancer receiving irinotecan. Irinotecan 210-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 21486535-1 2010 PURPOSE: Functional polymorphisms of the UGT1A1 gene, particularly the UGT1A1*28 variant, are associated with the severity of the bone marrow suppression in patients with metastatic colorectal cancer receiving irinotecan. Irinotecan 210-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 20358470-4 2010 We also explored the association between the UGT1A1*28 polymorphism and serum bilirubin concentrations and DNA damage and repair measures. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 20377135-0 2010 The UGT1A1*28 genotype and the toxicity of low-dose irinotecan in patients with advanced lung cancer. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 20377135-1 2010 The association between the UGT1A1*28 genotype and the severe toxicity of low-dose irinotecan has been controversial, and few studies have examined this association in patients with lung cancer. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 20377135-2 2010 The aim of this study was to assess the association between the UGT1A1*28 genotype and the severe toxicity of low-dose irinotecan in Japanese patients with lung cancer. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 20377135-11 2010 Therefore, low-dose irinotecan could be administered without reducing starting dose in patients with UGT1A1*28 genotype. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 20402064-4 2010 Thymine-adenine (TA) repeats in the promoter region of the UGT1A1 gene were investigated by polymerase chain reaction (PCR)-based non-radioactive DNA sequencing. Tantalum 17-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 19608554-0 2009 Prolonged neutropenia after irinotecan-based chemotherapy in a child with polymorphisms of UGT1A1 and SLCO1B1. Irinotecan 28-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 19608554-1 2009 Genetic polymorphisms of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and SLCO1B1 coding organic anion-transporter polypeptide 1B1, are independent risk factors known to increase irinotecan toxicity in adults. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-73 19608554-1 2009 Genetic polymorphisms of uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1), and SLCO1B1 coding organic anion-transporter polypeptide 1B1, are independent risk factors known to increase irinotecan toxicity in adults. Irinotecan 193-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 19608554-5 2009 To our knowledge, this is the first case report of combined genotyping of both UGT1A1 and SLCO1B1 in a child with severe irinotecan toxicity. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 20071295-2 2009 Its active metabolite, 7-ethyl-10 hydroxycamptothecin (SN-38) is glucuronidated by a uridine-diphosphoglucuronosyltransferase (UGT1A1) to form an inactive metabolite. Irinotecan 23-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 19415281-8 2009 UGT1A1 polymorphisms were associated with variability in irinotecan PK. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19747074-6 2009 Homeostatic feedback loops might not only include CYP1A1 but also Phase II enzymes such as UGT1A1 which controls the antioxidant AhR ligand bilirubin. Bilirubin 140-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 19672597-0 2009 UGT1A1 polymorphism and hyperbilirubinemia in a patient who received sorafenib. Sorafenib 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19672597-1 2009 PURPOSE: To report a single case of uridine glucuronosyltransferase 1A1 (UGT1A1) polymorphism and hyperbilirubinemia in a patient who received sorafenib. Sorafenib 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-71 19672597-1 2009 PURPOSE: To report a single case of uridine glucuronosyltransferase 1A1 (UGT1A1) polymorphism and hyperbilirubinemia in a patient who received sorafenib. Sorafenib 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19672597-8 2009 CONCLUSIONS: The isolated increase in serum bilirubin levels in our patient was probably due to sorafenib-induced UGT1A1 inhibition that manifested itself due both to the patient having one UGT1A1*28 allele and the presence of underlying liver disease. Bilirubin 44-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 19672597-8 2009 CONCLUSIONS: The isolated increase in serum bilirubin levels in our patient was probably due to sorafenib-induced UGT1A1 inhibition that manifested itself due both to the patient having one UGT1A1*28 allele and the presence of underlying liver disease. Bilirubin 44-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 19672597-8 2009 CONCLUSIONS: The isolated increase in serum bilirubin levels in our patient was probably due to sorafenib-induced UGT1A1 inhibition that manifested itself due both to the patient having one UGT1A1*28 allele and the presence of underlying liver disease. Sorafenib 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 19672597-8 2009 CONCLUSIONS: The isolated increase in serum bilirubin levels in our patient was probably due to sorafenib-induced UGT1A1 inhibition that manifested itself due both to the patient having one UGT1A1*28 allele and the presence of underlying liver disease. Sorafenib 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 19861894-8 2009 Formalin-fixed paraffin-embedded (FFPE) tissue was tested for UGT1A1 promoter variants by polymerase chain reaction and capillary electrophoresis. Formaldehyde 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 19861894-9 2009 To our knowledge, this is the first study to successfully apply UGT1A1 promoter genotyping to formalin-fixed paraffin-embedded tissue, which may facilitate more thorough examination of clinicopathologic correlations. Formaldehyde 94-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 19861894-9 2009 To our knowledge, this is the first study to successfully apply UGT1A1 promoter genotyping to formalin-fixed paraffin-embedded tissue, which may facilitate more thorough examination of clinicopathologic correlations. Paraffin 109-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 20071295-2 2009 Its active metabolite, 7-ethyl-10 hydroxycamptothecin (SN-38) is glucuronidated by a uridine-diphosphoglucuronosyltransferase (UGT1A1) to form an inactive metabolite. Irinotecan 55-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 20071295-3 2009 It was supposed that patients with the UGT1A1*28 polymorphism would have a greater prevalence of elevated pretreatment serum bilirubin levels and higher toxicity. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 19710077-11 2009 Indirect bilirubin increased 1.6- to 2.8-fold more in subjects with UGT1A1 *28/*28 versus *1/*28 or *1/*1. Bilirubin 9-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 19732760-0 2009 Haplotypes in the UGT1A1 gene and their role as genetic determinants of bilirubin concentration in healthy German volunteers. Bilirubin 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 19732760-1 2009 BACKGROUND: Genetic variations of UDP-glucuronyltransferase 1A1 (UGT1A1) influence the concentration of serum bilirubin. Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-63 19732760-1 2009 BACKGROUND: Genetic variations of UDP-glucuronyltransferase 1A1 (UGT1A1) influence the concentration of serum bilirubin. Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 19732760-2 2009 We investigated the association of four common polymorphisms including UGT1A1-53(TA)(n), and common haplotypes of the UGT1A1 gene with bilirubin levels in 218 Caucasian volunteers. Bilirubin 135-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 19732760-9 2009 Male sex, UGT1A1-53(TA)(6/7) and the c.-3279GG variant were significantly associated with higher bilirubin concentrations. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 19732760-10 2009 CONCLUSIONS: Two UGT1A1 promoter polymorphisms (-53(TA)(6/7) and c.-3279T>G) and a common haplotype of the UGT1A1 gene are associated with serum bilirubin concentrations in Caucasians. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 19732760-10 2009 CONCLUSIONS: Two UGT1A1 promoter polymorphisms (-53(TA)(6/7) and c.-3279T>G) and a common haplotype of the UGT1A1 gene are associated with serum bilirubin concentrations in Caucasians. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 19948621-2 2009 Variants in the UGT1A1 gene contribute to increased bilirubin levels, and bilirubin can act as an antioxidant. Bilirubin 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 19948621-2 2009 Variants in the UGT1A1 gene contribute to increased bilirubin levels, and bilirubin can act as an antioxidant. Bilirubin 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 19770637-0 2009 UGT1A1 genotyping: a predictor of irinotecan-associated side effects and drug efficacy? Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 19770637-3 2009 SN-38 is inactivated by uridine disphosphate glucuronosyl transferase 1 (UGT1A1) into the inactive compound SN-38G, which is excreted with the bile.This review concentrates on a critical evaluation of UGT1A1 gene polymorphism as a predictor of toxicity and treatment efficacy in patients who received irinotecan for metastatic colorectal cancer. Irinotecan 301-311 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19811694-6 2009 Resveratrol ranging from 1 to 5 microm could significantly suppress the expressions of cytochrome P450 (CYP) 1A1, CYP1B1 and UDP-glucuronosyltransferase (UGT) 1A1 induced by 7,12-dimethylbenz[a]anthracene (DMBA). Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-162 19811694-6 2009 Resveratrol ranging from 1 to 5 microm could significantly suppress the expressions of cytochrome P450 (CYP) 1A1, CYP1B1 and UDP-glucuronosyltransferase (UGT) 1A1 induced by 7,12-dimethylbenz[a]anthracene (DMBA). 7,12-dimethylbenz[a 174-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-162 19811694-6 2009 Resveratrol ranging from 1 to 5 microm could significantly suppress the expressions of cytochrome P450 (CYP) 1A1, CYP1B1 and UDP-glucuronosyltransferase (UGT) 1A1 induced by 7,12-dimethylbenz[a]anthracene (DMBA). anthracene 194-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-162 19710077-15 2009 The wild-type ABCB1 CGC haplotype was associated with slower CL/F and the UGT1A1 *28 genotype was associated with increased bilirubin. Bilirubin 124-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 19710077-16 2009 Thus, CYP3A5, ABCB1 and UGT1A1 polymorphisms are associated with atazanavir pharmacokinetics and pharmacodynamics in vivo. Atazanavir Sulfate 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 19732436-9 2009 siRNA treatment against UGT1A also showed an increase in MTX sensitivity. Methotrexate 57-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-29 19859999-0 2009 UGT1A1 gene polymorphism: impact on toxicity and efficacy of irinotecan-based regimens in metastatic colorectal cancer. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19859999-1 2009 AIM: To investigate the correlation between uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) gene polymorphisms and irinotecan-associated side effects and parameters of drug efficacy in patients with metastatic colorectal cancer (mCRC) receiving a low-dose weekly irinotecan chemotherapeutic regimen. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-92 19859999-1 2009 AIM: To investigate the correlation between uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) gene polymorphisms and irinotecan-associated side effects and parameters of drug efficacy in patients with metastatic colorectal cancer (mCRC) receiving a low-dose weekly irinotecan chemotherapeutic regimen. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 19859999-2 2009 METHODS: Genotypes were retrospectively evaluated by gene scan analysis on the ABI 310 sequencer of the TATAA box in the promoter region of the UGT1A1 gene in blood samples from 105 patients who had received 1st line irinotecan-based chemotherapy for mCRC. Irinotecan 217-227 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 19859999-7 2009 CONCLUSION: This analysis demonstrates the non-significant influence of the UGT1A1 gene polymorphism on efficacy and rate of irinotecan-associated toxicity in mCRC patients receiving low-dose irinotecan based chemotherapy. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 19389676-0 2009 Conditional linkage and genome-wide association studies identify UGT1A1 as a major gene for anti-atherogenic serum bilirubin levels--the Framingham Heart Study. Bilirubin 115-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 19389676-10 2009 CONCLUSIONS: Our studies suggest that UGT1A1 may be the major gene with strong effects on bilirubin levels and the TA-repeat polymorphism might be the key polymorphism within the gene controlling bilirubin levels. Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 19389676-10 2009 CONCLUSIONS: Our studies suggest that UGT1A1 may be the major gene with strong effects on bilirubin levels and the TA-repeat polymorphism might be the key polymorphism within the gene controlling bilirubin levels. Bilirubin 196-205 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 19487247-0 2009 Influence of N-terminal domain histidine and proline residues on the substrate selectivities of human UDP-glucuronosyltransferase 1A1, 1A6, 1A9, 2B7, and 2B10. Histidine 31-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-133 19517472-1 2009 BACKGROUND: The objective of this study was to examine the cost effectiveness of using a pharmacogenetic test for uridine diphosphate glycosyltransferase 1A1*28 (UGT1A1*28) variant homozygosity before administering irinotecan to patients with metastatic colorectal cancer. Irinotecan 215-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 19517472-4 2009 With genetic testing, irinotecan dosage was reduced 25% in homozygotes with the UGT1A1*28 variant allele. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 19487247-0 2009 Influence of N-terminal domain histidine and proline residues on the substrate selectivities of human UDP-glucuronosyltransferase 1A1, 1A6, 1A9, 2B7, and 2B10. Proline 45-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-133 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. Naproxen 256-264 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19487247-1 2009 An N-terminal domain histidine [corresponding to position 39 of UDP-glucuronosyltransferase (UGT) 1A1] is conserved in all UGT1A and UGT2B subfamily proteins except UGT1A4 (Pro-40) and UGT2B10 (Leu-34). Histidine 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-101 19487247-6 2009 Whereas UGT1A1(H39P), UGT1A6(H38P), and UGT1A9(H37P) lacked the ability to metabolize 4MU, 1NP, and naproxen, all glucuronidated LTG. Hymecromone 86-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 19487247-6 2009 Whereas UGT1A1(H39P), UGT1A6(H38P), and UGT1A9(H37P) lacked the ability to metabolize 4MU, 1NP, and naproxen, all glucuronidated LTG. 1-naphthol 91-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 19487247-6 2009 Whereas UGT1A1(H39P), UGT1A6(H38P), and UGT1A9(H37P) lacked the ability to metabolize 4MU, 1NP, and naproxen, all glucuronidated LTG. Naproxen 100-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 19541828-0 2009 Genetic polymorphisms in the TATA box and upstream phenobarbital-responsive enhancer module of the UGT1A1 promoter have combined effects on UDP-glucuronosyltransferase 1A1 transcription mediated by constitutive androstane receptor, pregnane X receptor, or glucocorticoid receptor in human liver. Phenobarbital 51-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 19541828-0 2009 Genetic polymorphisms in the TATA box and upstream phenobarbital-responsive enhancer module of the UGT1A1 promoter have combined effects on UDP-glucuronosyltransferase 1A1 transcription mediated by constitutive androstane receptor, pregnane X receptor, or glucocorticoid receptor in human liver. Phenobarbital 51-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-171 19487247-6 2009 Whereas UGT1A1(H39P), UGT1A6(H38P), and UGT1A9(H37P) lacked the ability to metabolize 4MU, 1NP, and naproxen, all glucuronidated LTG. Lamotrigine 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 19487247-1 2009 An N-terminal domain histidine [corresponding to position 39 of UDP-glucuronosyltransferase (UGT) 1A1] is conserved in all UGT1A and UGT2B subfamily proteins except UGT1A4 (Pro-40) and UGT2B10 (Leu-34). Leucine 194-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-101 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. Histidine 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. Proline 95-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19541828-2 2009 The purpose of this study was to determine whether the genetic polymorphisms in the RNA polymerase II core promoter and the upstream phenobarbital-responsive element module (PBREM) of the UGT1A1 promoter have combined effects on UGT1A1 transcription mediated by the transcription factors. Phenobarbital 133-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 19541828-2 2009 The purpose of this study was to determine whether the genetic polymorphisms in the RNA polymerase II core promoter and the upstream phenobarbital-responsive element module (PBREM) of the UGT1A1 promoter have combined effects on UGT1A1 transcription mediated by the transcription factors. Phenobarbital 133-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 229-235 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. Leucine 107-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. Histidine 150-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19487247-5 2009 The conserved N-terminal domain histidine of UGT1A1, UGT1A6, UGT1A9, and UGT2B7 was mutated to proline and leucine 34 of UGT2B10 was substituted with histidine, and the capacity of the wild-type and mutant proteins to glucuronidate 4MU, 1NP, LTG, TFP, and naproxen was characterized. glucuronidate 218-231 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19486934-5 2009 It has been reported that UDP-glucuronosyltransferase (UGT) 1A1, UGT1A8, UGT1A9, and UGT1A10 are enzymes for raloxifene glucuronidation, and UGT1A8 and UGT1A10 are absent in the human liver, whereas UGT1A1, UGT1A8, UGT1A9, and UGT1A10 are present in the human intestine. Raloxifene Hydrochloride 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-63 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 31-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 43-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-1 2009 SN-38, an active metabolite of irinotecan (CPT-11), is glucuronidated into SN-38G mainly by UGT1A1, during detoxification. Irinotecan 75-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19755821-2 2009 However, significant interindividual pharmacokinetic variability in SN-38 is caused by factors, including inherited predispositions that may affect the function and expression of UGT1A1. Irinotecan 68-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 19755821-4 2009 Similar to SN-38, bilirubin is excreted into bile after being glucuronidated by UGT1A1. Bilirubin 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 20084161-8 2009 The US Food and Drug Administration has approved the UDP-glucuronosyltransferase 1A1 test in patients treated with irinotecan, and additional approval of newer tests is anticipated. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-84 19545173-2 2009 The results indicated that these isoflavones are metabolized most rapidly at three different concentrations by one of these four UGT isoforms: UGT1A1, UGT1A8, UGT1A9 and UGT1A10. Isoflavones 33-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 19486253-1 2009 BACKGROUND AND AIMS: The gene product of the uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is crucial to bilirubin metabolism. Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-93 19486253-1 2009 BACKGROUND AND AIMS: The gene product of the uridine diphosphate glucuronosyl transferase 1A1 (UGT1A1) is crucial to bilirubin metabolism. Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 19482841-6 2009 The effects of clinical factors and variants of the UGT1A1 gene on serum bilirubin levels were determined. Bilirubin 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 87-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-4 2009 Irinotecan is also metabolized by carboxylesterase to its principal active metabolite, SN-38, which is subsequently glucuronidated by UGT1As to form the inactive compound SN-38G. Irinotecan 171-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-138 20719167-5 2009 UGT1A1(*)28 and UGT1A1(*)6 polymorphisms were useful for predicting severe toxicity with NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 20719167-5 2009 UGT1A1(*)28 and UGT1A1(*)6 polymorphisms were useful for predicting severe toxicity with NSCLC patients treated with irinotecan-based chemotherapy. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 19486934-5 2009 It has been reported that UDP-glucuronosyltransferase (UGT) 1A1, UGT1A8, UGT1A9, and UGT1A10 are enzymes for raloxifene glucuronidation, and UGT1A8 and UGT1A10 are absent in the human liver, whereas UGT1A1, UGT1A8, UGT1A9, and UGT1A10 are present in the human intestine. Raloxifene Hydrochloride 109-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 19372226-8 2009 Incubations with recombinant human UDP-glucuronosyltransferases (UGTs) and inhibition by the UGT1A4 and UGT1A1 substrates/inhibitors imipramine and bilirubin suggested that UGT1A4 is the major UGT isozyme catalyzing the N-glucuronidation of motesanib, with a minor contribution from UGT1A1. Imipramine 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 19406951-0 2009 Characterizing the effects of common UDP glucuronosyltransferase (UGT) 1A6 and UGT1A1 polymorphisms on cis- and trans-resveratrol glucuronidation. Resveratrol 112-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 19339377-9 2009 Further oxidation to THC-COOH surprisingly leads to a loss in metabolism by UGT1A9 and UGT1A10, while creating a substrate recognized by UGT1A1 and UGT1A3. Dronabinol 21-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 19414484-3 2009 Meta-analysis showed strong replication for a genetic influence on serum bilirubin levels of the UGT1A1 locus (P < 5 x 10(-324)) and a 12p12.2 locus. Bilirubin 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 19414484-6 2009 The top variants in UGT1A1 and SLCO1B1 explain approximately 18.0 and approximately 1.0% of the variation in total serum bilirubin levels, respectively. Bilirubin 121-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 19414484-8 2009 In one of the largest genetic studies of bilirubin to date (n = 9464), we confirm the substantial genetic influence of UGT1A1 variants, consistent with past linkage and association studies, and additionally provide strong evidence of a role for allelic variation in SLCO1B1. Bilirubin 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 19414484-10 2009 In analyses for association with gallbladder disease or gallstones, top bilirubin SNPs in UGT1A1 and SLCO1B1 were not associated. Bilirubin 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 19419973-5 2009 In addition to the two known loci previously involved in the regulation of bilirubin levels, UGT1A1 (P = 6.2 x 10(-62)) and G6PD (P = 2.5 x 10(-8)), we observed a strong association on chromosome 12 within the SLCO1B3 gene (P = 3.9 x 10(-9)). Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 19397531-5 2009 In the hyperbilirubinemic group, 23 of 91 (25.3%) infants demonstrated variant of UGT1A1 at nucleotides (nt) 211 as compared to 6 of 86 (7%) in the control group (p = 0.001). Nucleotides 54-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 19841506-4 2009 Of note, both patients recovered from their initial unconjugated hyperbilirubinemia despite continuation of ribavirin therapy, which indicates that compensatory mechanisms leading to a normalization of UGT1A1 activity are likely. Ribavirin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 202-208 19339377-9 2009 Further oxidation to THC-COOH surprisingly leads to a loss in metabolism by UGT1A9 and UGT1A10, while creating a substrate recognized by UGT1A1 and UGT1A3. Carbonic Acid 25-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 19372226-8 2009 Incubations with recombinant human UDP-glucuronosyltransferases (UGTs) and inhibition by the UGT1A4 and UGT1A1 substrates/inhibitors imipramine and bilirubin suggested that UGT1A4 is the major UGT isozyme catalyzing the N-glucuronidation of motesanib, with a minor contribution from UGT1A1. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 19372226-8 2009 Incubations with recombinant human UDP-glucuronosyltransferases (UGTs) and inhibition by the UGT1A4 and UGT1A1 substrates/inhibitors imipramine and bilirubin suggested that UGT1A4 is the major UGT isozyme catalyzing the N-glucuronidation of motesanib, with a minor contribution from UGT1A1. Nitrogen 220-221 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 19372226-8 2009 Incubations with recombinant human UDP-glucuronosyltransferases (UGTs) and inhibition by the UGT1A4 and UGT1A1 substrates/inhibitors imipramine and bilirubin suggested that UGT1A4 is the major UGT isozyme catalyzing the N-glucuronidation of motesanib, with a minor contribution from UGT1A1. motesanib diphosphate 241-250 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Sterols 76-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 19620808-1 2009 Severe neutropenia attributable to irinotecan hydrochloride (CPT-11) is reportedly associated with gene polymorphism of UGT1A1 related to its metabolism. Irinotecan 35-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 19620808-1 2009 Severe neutropenia attributable to irinotecan hydrochloride (CPT-11) is reportedly associated with gene polymorphism of UGT1A1 related to its metabolism. Irinotecan 61-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 19620808-9 2009 Nine days following administration of the first course, we found neutropenia of grade 3 and a fever of 38 degrees C. Analysis of the UGT1A1 gene polymorphism after symptom improvement revealed UGT1A1(*)28 homozygosity in both cases, which suggests that when FOLFIRI is conducted on a patient with homozygous UGT1A1(*)28, it is necessary to pay attention to neutropenia even with a CPT-11 dosage of 120 mg/m(2). Irinotecan 381-387 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 193-199 19513514-14 2009 However, significant genotypic variation of UGT1A1, which was assumed to affect irinotecan toxicity, was not observed to affect RR, toxicity or survival. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Sterols 76-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 220-226 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Steroids 87-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 209-215 19409404-6 2009 Differentially expressed genes included the Phase I xenobiotic, fatty acid, sterol and steroid metabolism genes Cyp2b2 and CYP2B6, Cyp3a1 and CYP3A4, and Cyp4a22 and CYP4A11; Phase II conjugation enzyme genes Ugt1a1 and UGT1A1; and Phase III ABC transporter genes Abcb1 and ABCB1. Steroids 87-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 220-226 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 65-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19279563-6 2009 Plasma concentrations of raltegravir are modestly higher in individuals with the UGT1A1*28/*28 genotype than in those with the UGT1A1*1/*1 genotype. Raltegravir Potassium 25-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 19349540-1 2009 PURPOSE: We aim to identify genetic variation, in addition to the UGT1A1*28 polymorphism, that can explain the variability in irinotecan (CPT-11) pharmacokinetics and neutropenia in cancer patients. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 19349540-1 2009 PURPOSE: We aim to identify genetic variation, in addition to the UGT1A1*28 polymorphism, that can explain the variability in irinotecan (CPT-11) pharmacokinetics and neutropenia in cancer patients. Irinotecan 138-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 19349540-7 2009 Almost 30% of the variability in SN-38 (the active metabolite of CPT-11) AUC is explained by ABCC1 1684T>C, ABCB1 IVS9 -44A>G, and UGT1A1*93 (P = .004). Irinotecan 33-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19279563-0 2009 Pharmacokinetics of raltegravir in individuals with UGT1A1 polymorphisms. Raltegravir Potassium 20-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 19279563-1 2009 Raltegravir is a human immunodeficiency virus-1 (HIV-1) integrase strand transfer inhibitor metabolized by glucuronidation via UDP-glucuronosyltransferase 1A1 (UGT1A1). Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 19279563-4 2009 The geometric mean ratio (GMR) (UGT1A1*28/*28 to UGT1A1*1/*1) and 90% confidence interval (CI) were 1.41 (0.96, 2.09) for raltegravir area under the concentration-time curve (AUC(0-infinity)), 1.40 (0.86, 2.28) for maximum plasma concentration (C(max)), and 1.91 (1.43, 2.55) for concentration at the 12-h time point (C(12 h)). Raltegravir Potassium 122-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 19279563-4 2009 The geometric mean ratio (GMR) (UGT1A1*28/*28 to UGT1A1*1/*1) and 90% confidence interval (CI) were 1.41 (0.96, 2.09) for raltegravir area under the concentration-time curve (AUC(0-infinity)), 1.40 (0.86, 2.28) for maximum plasma concentration (C(max)), and 1.91 (1.43, 2.55) for concentration at the 12-h time point (C(12 h)). Raltegravir Potassium 122-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 19279563-6 2009 Plasma concentrations of raltegravir are modestly higher in individuals with the UGT1A1*28/*28 genotype than in those with the UGT1A1*1/*1 genotype. Raltegravir Potassium 25-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 19251824-5 2009 In HepG2 and LS180 cells stably transduced with PXR.1, PXR target genes (CYP3A4, MDR1, CYP2B6, and UGT1A1) were higher than mock-transduced cells in the absence of ligand and were further induced in the presence of rifampin. Rifampin 215-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 19343046-7 2009 Results of linear multiple regression analysis on serum total bilirubin levels followed by analysis of variance showed that at least 13% of the variance in serum total bilirubin levels could be explained by three haplotype-tagging SNPs in the UGT1A1 gene. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 19336732-0 2009 Cruciferous vegetable feeding alters UGT1A1 activity: diet- and genotype-dependent changes in serum bilirubin in a controlled feeding trial. Bilirubin 100-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 19343046-2 2009 On analysis, we observed a strong association between five SNPs within the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene and serum total bilirubin levels (minimum P-value in Mann-Whitney test=1.82 x 10(10)). Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-122 19343046-2 2009 On analysis, we observed a strong association between five SNPs within the uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) gene and serum total bilirubin levels (minimum P-value in Mann-Whitney test=1.82 x 10(10)). Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 19343046-3 2009 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid, thus enhancing bilirubin elimination. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19343046-3 2009 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid, thus enhancing bilirubin elimination. Glucuronic Acid 51-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19343046-3 2009 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid, thus enhancing bilirubin elimination. Bilirubin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19343046-7 2009 Results of linear multiple regression analysis on serum total bilirubin levels followed by analysis of variance showed that at least 13% of the variance in serum total bilirubin levels could be explained by three haplotype-tagging SNPs in the UGT1A1 gene. Bilirubin 168-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 19364970-1 2009 PURPOSE: UGT1A1*28 is considered the main pharmacogenetic predictor of the toxicity outcome of irinotecan-treated patients. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 19223645-2 2009 Raltegravir is primarily cleared by hepatic metabolism via glucuronidation (via UDP glucuronosyltransferase 1A1), with a minor component of elimination occurring via the renal pathway. Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-111 18998132-0 2009 Life-threatening toxicities in a patient with UGT1A1*6/*28 and SLCO1B1*15/*15 genotypes after irinotecan-based chemotherapy. Irinotecan 94-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18998132-6 2009 CONCLUSION: The severe toxicities in this patient are attributable to the extensive accumulation of SN-38, which may result from a synergistic or additive effect of low metabolic (UGT1A1*6/*28) and transport (SLCO1B1*15/*15) capabilities. Irinotecan 100-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 19303655-4 2009 Indinavir (IDV) can lead to hyperbilirubinemia in Gilbert"s syndrome (UGT1A1*28), which does not explain interindividual severity differences and may thus involve additional UGT1A variants. Indinavir 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 19364970-10 2009 CONCLUSION: We propose that UGT1A variants additional to UGT1A1*28 might improve the prediction of the outcome of colorectal cancer patients treated with FOLFIRI. FOLFIRI regimen 154-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 19207584-0 2009 UGT1A1 gene polymorphism as a potential factor inducing iron overload in the pathogenesis of type 1 hereditary hemochromatosis. Iron 56-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19207584-6 2009 Results High incidence and a significant correlation of UGT1A1 gene mutations with increased serum bilirubin level and lower grades of liver tissue inflammatory activity were observed in study participants. Bilirubin 99-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 19207584-9 2009 Conclusions UGT1A1 gene polymorphism and as its consequence of high serum bilirubin level may promote iron accumulation in hemochromatosis patients by reducing the activity of inflammation. Bilirubin 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 19207584-9 2009 Conclusions UGT1A1 gene polymorphism and as its consequence of high serum bilirubin level may promote iron accumulation in hemochromatosis patients by reducing the activity of inflammation. Iron 102-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 19450125-0 2009 UGT1A1*28 genotype predicts gastrointestinal toxicity in patients treated with intermediate-dose irinotecan. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19450125-1 2009 AIMS: Variants in UGT1A1 have previously been associated with toxicity from irinotecan chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 19450125-2 2009 We conducted a pragmatic prospective cohort study to establish the relevance of UGT1A1 variants in the prediction of severe diarrhea and neutropenia in patients with colorectal cancer receiving irinotecan in a routine clinical setting. Irinotecan 194-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 19371317-0 2009 Effects of UGT1A1*28 polymorphism on raloxifene pharmacokinetics and pharmacodynamics. Raloxifene Hydrochloride 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 19371317-2 2009 Bilirubin is a typical UGT1A1 substrate. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 19371317-3 2009 Based on these facts, we postulated a hypothesis that UGT1A1 is the key enzyme for metabolic clearance of raloxifene and that the common UGT1A1*28 polymorphism significantly contributes to the large pharmacokinetic variability of raloxifene. Raloxifene Hydrochloride 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 19371317-3 2009 Based on these facts, we postulated a hypothesis that UGT1A1 is the key enzyme for metabolic clearance of raloxifene and that the common UGT1A1*28 polymorphism significantly contributes to the large pharmacokinetic variability of raloxifene. Raloxifene Hydrochloride 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19371317-3 2009 Based on these facts, we postulated a hypothesis that UGT1A1 is the key enzyme for metabolic clearance of raloxifene and that the common UGT1A1*28 polymorphism significantly contributes to the large pharmacokinetic variability of raloxifene. Raloxifene Hydrochloride 230-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 19371317-10 2009 CONCLUSIONS: In this study it is shown that a relatively common UGT1A1*28 polymorphism may considerably influence raloxifene pharmacokinetics and pharmacodynamics. Raloxifene Hydrochloride 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 19336732-2 2009 UGT1A1 glucuronidates bilirubin, estrogens, and several dietary carcinogens. Bilirubin 22-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19034627-6 2009 Notably, the flavonoids induced the expression of UGT1A1 and CYP1A2 in HepG2 cells but not in HPH. Flavonoids 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 19390945-0 2009 UGT1A1*6 polymorphism is most predictive of severe neutropenia induced by irinotecan in Japanese cancer patients. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19390945-1 2009 BACKGROUND: Gene polymorphisms of the UDP-glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) contribute to individual variations in adverse events among patients administered irinotecan, and the distribution of the polymorphisms shows large interethnic differences. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-90 19390945-1 2009 BACKGROUND: Gene polymorphisms of the UDP-glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) contribute to individual variations in adverse events among patients administered irinotecan, and the distribution of the polymorphisms shows large interethnic differences. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 19390945-3 2009 In the present study, we evaluated the association of genetic polymorphisms of UGT1A1 and SLCO1B1 with irinotecanrelated neutropenia in Japanese cancer patients. irinotecanrelated 103-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 19390945-12 2009 CONCLUSION: These findings suggest that the UGT1A1*6 polymorphism is a potential predictor of severe neutropenia caused by irinotecan in Japanese cancer patients. Irinotecan 123-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 19601871-2 2009 The involvement of UGT 1A1, 1A3 and 2B7 in buprenorpine and 1A3 in norbuprenorphine glucuronidation were confirmed. buprenorpine 43-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-26 19601871-2 2009 The involvement of UGT 1A1, 1A3 and 2B7 in buprenorpine and 1A3 in norbuprenorphine glucuronidation were confirmed. norbuprenorphine 67-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-26 19034627-4 2009 RESULTS: In human primary hepatocytes, real-time PCR analysis showed induction of CYP2B6, CYP3A4, UGT1A1, MDR1, and MRP2 by EGb 761, ginkgolide A (GA) and ginkgolide B (GB), but not by bilobalide (BB) or the flavonoids (quercetin, kaempferol and tamarixetin) of GBE. BDBM50323769 124-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 19268007-0 2009 Does bilirubin level correspond to interaction of c.-3279T>G and A(TA)7TAA variants in UGT1A1 gene? Bilirubin 5-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 19141701-0 2009 Citrus fruit intake is associated with lower serum bilirubin concentration among women with the UGT1A1*28 polymorphism. Bilirubin 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 19141701-1 2009 UDP-glucuronosyltransferase (UGT) 1A1 glucuronidates bilirubin, estrogens, and xenobiotic compounds. Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 19141701-2 2009 The UGT1A1*28 polymorphism results in lower promoter activity due to 7 thymine-adenine (TA) repeats rather than the more common 6 TA repeats. 7 thymine-adenine 69-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19141701-2 2009 The UGT1A1*28 polymorphism results in lower promoter activity due to 7 thymine-adenine (TA) repeats rather than the more common 6 TA repeats. Tantalum 88-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19141701-3 2009 Previously, we showed that serum bilirubin, a marker of UGT1A1 activity, was lower among individuals homozygous for the UGT1A1*28 polymorphism (7/7) when randomized to a high fruit and vegetable (F&V) diet, whereas there was no effect in individuals with the wild-type (6/6) and heterozygous (6/7) genotypes. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 19141701-3 2009 Previously, we showed that serum bilirubin, a marker of UGT1A1 activity, was lower among individuals homozygous for the UGT1A1*28 polymorphism (7/7) when randomized to a high fruit and vegetable (F&V) diet, whereas there was no effect in individuals with the wild-type (6/6) and heterozygous (6/7) genotypes. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-126 19268007-1 2009 Promoter variants c.-3279T>G and A(TA)7TAA show decreased level of expression of UDP-glucuronosyl transferase 1A1 (UGT1A1) and consequently reduced activity of the enzyme catalyzing glucuronidation of bilirubin in hepatocytes. Bilirubin 204-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-116 19268007-1 2009 Promoter variants c.-3279T>G and A(TA)7TAA show decreased level of expression of UDP-glucuronosyl transferase 1A1 (UGT1A1) and consequently reduced activity of the enzyme catalyzing glucuronidation of bilirubin in hepatocytes. Bilirubin 204-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-124 18685565-0 2009 Phase I/II pharmacokinetic and pharmacogenomic study of UGT1A1 polymorphism in elderly patients with advanced non-small cell lung cancer treated with irinotecan. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 18685565-9 2009 An analysis of the influence of UGT1A1*28 and *6 polymorphisms provides useful information for the prediction of CPT-11-related hematological toxicity. Irinotecan 113-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 19571434-10 2009 In the present study, Kampo was clarified to inhibit beta-estradiol and SN-38 glucuronidation mainly catalyzed by UGT1A1. Estradiol 53-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 19414151-4 2009 UGT1A1*28 leads to reduced conjugation of SN-38, the active metabolite of irinotecan, resulting in an increased rate of adverse effects, especially neutropenia. Irinotecan 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19414151-4 2009 UGT1A1*28 leads to reduced conjugation of SN-38, the active metabolite of irinotecan, resulting in an increased rate of adverse effects, especially neutropenia. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18981166-0 2009 Close association of UGT1A9 IVS1+399C>T with UGT1A1*28, *6, or *60 haplotype and its apparent influence on 7-ethyl-10-hydroxycamptothecin (SN-38) glucuronidation in Japanese. Irinotecan 142-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 68-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 100-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-1 2009 The anticancer prodrug, irinotecan, is converted to its active form 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterases, and SN-38 is inactivated by UDP-glucuronosyltransferase (UGT)1A1-mediated glucuronidation. Irinotecan 133-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-193 18981166-11 2009 Thus, at least in Japanese populations, influence of I399C>T on SN-38 glucuronidation is attributable to its close association with either UGT1A1*6, *28, or *60. Irinotecan 67-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 19571434-10 2009 In the present study, Kampo was clarified to inhibit beta-estradiol and SN-38 glucuronidation mainly catalyzed by UGT1A1. Irinotecan 72-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 20045987-9 2009 These results suggest that glycyrrhetinic acid glucuronidation is primarily mediated by UGT1A1, 1A3, 2B4 and 2B7. Glycyrrhetinic Acid 27-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 18221820-5 2009 The UGT1A1*6/*6, UGT1A9*1/*1 or *1/*22, and SLCO1B1 521TC or CC genotypes, and female-gender were predictive for higher AUC(SN-38) in multivariate analysis. Irinotecan 124-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19125128-0 2009 Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Irinotecan 158-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 19125128-8 2009 CLINICAL VALIDITY: The EWG found adequate evidence of a significant association between UGT1A1 genotype and the incidence of severe neutropenia at standard doses of irinotecan. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 19125128-16 2009 Further rigorous evaluation of UGT1A1 genotyping using current and promising irinotecan treatment protocols is warranted. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 19125129-0 2009 Can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 19125129-2 2009 This evidence-based review addresses the question of whether testing for UGT1A1 mutations in patients with metastatic colorectal cancer treated with irinotecan leads to improvement in outcomes (e.g., irinotecan toxicity, response to treatment, morbidity, and mortality), when compared with no testing. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19125129-2 2009 This evidence-based review addresses the question of whether testing for UGT1A1 mutations in patients with metastatic colorectal cancer treated with irinotecan leads to improvement in outcomes (e.g., irinotecan toxicity, response to treatment, morbidity, and mortality), when compared with no testing. Irinotecan 200-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 19125129-16 2009 Given the large number of colorectal cancer cases diagnosed each year, a randomized controlled trial of the effects of irinotecan dose modifications in patients with colorectal cancer based on their UGT1A1 genotype is feasible and could clarify the tradeoffs between possible reductions in severe neutropenia and improved tumor response and/or survival in patients with various UGT1A1 genotypes. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 199-205 19125129-16 2009 Given the large number of colorectal cancer cases diagnosed each year, a randomized controlled trial of the effects of irinotecan dose modifications in patients with colorectal cancer based on their UGT1A1 genotype is feasible and could clarify the tradeoffs between possible reductions in severe neutropenia and improved tumor response and/or survival in patients with various UGT1A1 genotypes. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 378-384 19356098-0 2008 Induction of human UGT1A1 by bilirubin through AhR dependent pathway. Bilirubin 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 19325249-1 2009 BACKGROUND: The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) enzyme is responsible for conjugation of the bilirubin in the liver as well as for drug metabolism. Bilirubin 118-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-63 19325249-1 2009 BACKGROUND: The uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) enzyme is responsible for conjugation of the bilirubin in the liver as well as for drug metabolism. Bilirubin 118-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 19299905-0 2009 Clinical significance of UDP-glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers: a prospective multi-institutional study. Irinotecan 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-56 19299905-0 2009 Clinical significance of UDP-glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers: a prospective multi-institutional study. Cisplatin 122-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-56 19299905-1 2009 BACKGROUND: To investigate the effects of UDP-glucuronosyltransferase 1A1 (UGT1A1) *28, *6 and *27 in patients with gynecologic cancer who received chemotherapy with irinotecan and cisplatin. Irinotecan 166-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-73 19299905-9 2009 CONCLUSION: In addition to UGT1A1*28, UGT1A1*6 might also be a key candidate to determine the dose of combination chemotherapy with irinotecan and cisplatin. Irinotecan 132-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 19299905-9 2009 CONCLUSION: In addition to UGT1A1*28, UGT1A1*6 might also be a key candidate to determine the dose of combination chemotherapy with irinotecan and cisplatin. Cisplatin 147-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 18953066-0 2008 Genetic testing for UGT1A1*28 and *6 in Japanese patients who receive irinotecan chemotherapy. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 19059062-3 2008 Irinotecan toxicity in patients who have colorectal cancer has been linked to reduced activity of uridine diphosphate-glucuronyltransferase 1A1 (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-143 19059062-3 2008 Irinotecan toxicity in patients who have colorectal cancer has been linked to reduced activity of uridine diphosphate-glucuronyltransferase 1A1 (UGT1A1). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 145-151 19356098-2 2008 In this report, we show findings of the induction of the reporter gene (-3475/+14) of UGT1A1 in HepG2 cells by bilirubin at 50 microM, 100 microM, with human aryl hydrocarbon receptor (hAhR). Bilirubin 111-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 19712005-3 2009 METHODS: Here, we assess the genome of 469 individuals from Sao Miguel Island (Azores, Portugal) in order to determine the frequencies of polymorphisms and haplotypes in UGT1A1, UGT1A6, and UGT1A7, the co-occurrence of reduced enzyme activity UGT1A variants related to irinotecan toxicity, and to calculate the extent of linkage disequilibrium (LD) in the genomic region encompassing these genes. Irinotecan 269-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 19639031-6 2008 UGT1A1 genotype added approximately 8-9% during cycle 1 and from approximately 7% [mIFL regimen] to 26% [CapeIRI regimen] after cycle 1. capeiri 105-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18990428-3 2008 BPA glucuronidation was catalyzed by UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7 and UGT2B15 as well as by human liver microsomes. bisphenol A 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 18528434-1 2008 Atazanavir (ATV) is known to inhibit UGT1A1-mediated glucuronidation. Atazanavir Sulfate 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 18528434-1 2008 Atazanavir (ATV) is known to inhibit UGT1A1-mediated glucuronidation. Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 19356098-1 2008 UDP-glucuronosyltransferase1A1 (UGT1A1) plays a key role to conjugate bilirubin and preventing jaundice, but there is no report showing the induction of human UGT1A1 (UGT1A1) by bilirubin. Bilirubin 70-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 19356098-1 2008 UDP-glucuronosyltransferase1A1 (UGT1A1) plays a key role to conjugate bilirubin and preventing jaundice, but there is no report showing the induction of human UGT1A1 (UGT1A1) by bilirubin. Bilirubin 70-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 19356098-1 2008 UDP-glucuronosyltransferase1A1 (UGT1A1) plays a key role to conjugate bilirubin and preventing jaundice, but there is no report showing the induction of human UGT1A1 (UGT1A1) by bilirubin. Bilirubin 178-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 19356098-6 2008 These results indicate that the induction of UGT1A1 by bilirubin-AhR did not depend on the elevation of AhR but on ligand binding. Bilirubin 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 19356098-7 2008 From this result, we considered that high bilirubin in neonates must induce the elevation of UGT1A1 after birth to prevent jaundice, and bilirubin in adults also regulates the level of UGT1A1. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 19356098-7 2008 From this result, we considered that high bilirubin in neonates must induce the elevation of UGT1A1 after birth to prevent jaundice, and bilirubin in adults also regulates the level of UGT1A1. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 19356098-7 2008 From this result, we considered that high bilirubin in neonates must induce the elevation of UGT1A1 after birth to prevent jaundice, and bilirubin in adults also regulates the level of UGT1A1. Bilirubin 137-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 19356098-8 2008 This is the first report showing direct induction of UGT1A1 by a bilirubin through AhR pathway. Bilirubin 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 18558463-5 2008 Also, elevated bilirubin levels, due to liver impairment, conjugation disorders or UGT1A1 *28 genotype, have been associated with increased incidence of grades 3 intestinal toxicity and neutropenia. Bilirubin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 18335219-0 2008 Cisplatin plus weekly CPT-11/docetaxel in advanced esophagogastric cancer: a phase I study with pharmacogenetic assessment of XPD, XRCC3 and UGT1A1 polymorphisms. Cisplatin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 18335219-1 2008 PURPOSE: A multicenter phase I trial to establish the recommended dose of CPT-11/docetaxel plus cisplatin in advanced esophagogastric cancer patients and to correlate the efficacy and toxicity with genetic polymorphisms in DNA repair genes (XPD and XRCC3) and the UGT1A1 gene. Irinotecan 74-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 18790042-2 2008 UGT1A1 is the major gene influencing bilirubin concentrations. Bilirubin 37-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18790042-3 2008 Therefore, we investigated an association of bilirubin levels and two polymorphisms in the promoter of UGT1A1 (-53(TA-repeat) polymorphism and T-3279G) in 477 patients with premature, familial CAD and 619 age- and sex-matched controls. Bilirubin 45-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18335219-1 2008 PURPOSE: A multicenter phase I trial to establish the recommended dose of CPT-11/docetaxel plus cisplatin in advanced esophagogastric cancer patients and to correlate the efficacy and toxicity with genetic polymorphisms in DNA repair genes (XPD and XRCC3) and the UGT1A1 gene. Docetaxel 81-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 264-270 18558463-6 2008 UGT1A1 *28 homozygosity is strongly associated with irinotecan-induced neutropenia and polymorphisms in the transporting peptides ABCB1 and OATP1B1 have also been associated with gastrointestinal toxicity and irinotecan pharmacokinetics, respectively. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18558463-7 2008 In the irinotecan product label, it is advised to reduce the irinotecan starting dose for UGT1A1 *28 homozygotes. Irinotecan 7-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 18558463-7 2008 In the irinotecan product label, it is advised to reduce the irinotecan starting dose for UGT1A1 *28 homozygotes. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 18832463-2 2008 The basis of the disorder is a 70% reduction in bilirubin glucuronidation catalyzed by the UDP-glucuronosyltransferase 1A1 (UGT1A1), which, in Caucasians, is the result of a homozygous TA insertion into the promoter region of the UGT1A1 gene (UGT1A1*28). Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-122 18832463-2 2008 The basis of the disorder is a 70% reduction in bilirubin glucuronidation catalyzed by the UDP-glucuronosyltransferase 1A1 (UGT1A1), which, in Caucasians, is the result of a homozygous TA insertion into the promoter region of the UGT1A1 gene (UGT1A1*28). Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 18832463-2 2008 The basis of the disorder is a 70% reduction in bilirubin glucuronidation catalyzed by the UDP-glucuronosyltransferase 1A1 (UGT1A1), which, in Caucasians, is the result of a homozygous TA insertion into the promoter region of the UGT1A1 gene (UGT1A1*28). Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 18832463-2 2008 The basis of the disorder is a 70% reduction in bilirubin glucuronidation catalyzed by the UDP-glucuronosyltransferase 1A1 (UGT1A1), which, in Caucasians, is the result of a homozygous TA insertion into the promoter region of the UGT1A1 gene (UGT1A1*28). Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 18832463-3 2008 Homozygous carriers of UGT1A1*28 as well as those with additional UGT1A variants can suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. Irinotecan 104-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18430559-0 2008 Glucuronidation of the soyabean isoflavones genistein and daidzein by human liver is related to levels of UGT1A1 and UGT1A9 activity and alters isoflavone response in the MCF-7 human breast cancer cell line. Isoflavones 32-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 18832463-3 2008 Homozygous carriers of UGT1A1*28 as well as those with additional UGT1A variants can suffer from severe irinotecan toxicity or jaundice during treatment with the protease inhibitor atazanavir. Atazanavir Sulfate 181-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18430559-0 2008 Glucuronidation of the soyabean isoflavones genistein and daidzein by human liver is related to levels of UGT1A1 and UGT1A9 activity and alters isoflavone response in the MCF-7 human breast cancer cell line. Genistein 44-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 18634814-5 2008 Genistein, on the other hand, inhibited ST1E1 and UGT1A1 expressions, and led to 17beta-estradiol cellular retention. Genistein 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 18430559-0 2008 Glucuronidation of the soyabean isoflavones genistein and daidzein by human liver is related to levels of UGT1A1 and UGT1A9 activity and alters isoflavone response in the MCF-7 human breast cancer cell line. daidzein 58-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 18430559-0 2008 Glucuronidation of the soyabean isoflavones genistein and daidzein by human liver is related to levels of UGT1A1 and UGT1A9 activity and alters isoflavone response in the MCF-7 human breast cancer cell line. Isoflavones 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 18430559-2 2008 We observed a correlation between rates of metabolism of marker substrates of specific UGTs and rates of glucuronidation of genistein and daidzein in vitro by a panel of human liver microsomes, demonstrating that UGT1A1 and UGT1A9, but not UGT1A4, make a major contribution to the metabolism of these isoflavones by human liver. daidzein 138-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 213-219 18430559-2 2008 We observed a correlation between rates of metabolism of marker substrates of specific UGTs and rates of glucuronidation of genistein and daidzein in vitro by a panel of human liver microsomes, demonstrating that UGT1A1 and UGT1A9, but not UGT1A4, make a major contribution to the metabolism of these isoflavones by human liver. Isoflavones 301-312 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 213-219 18430559-3 2008 These findings were substantiated by observations that recombinant human UGT1A1 and UGT1A9, but not UGT1A4, catalysed the production of the major glucuronides of both genistein and daidzein in vitro. Glucuronides 146-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 18430559-3 2008 These findings were substantiated by observations that recombinant human UGT1A1 and UGT1A9, but not UGT1A4, catalysed the production of the major glucuronides of both genistein and daidzein in vitro. Genistein 167-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 18430559-3 2008 These findings were substantiated by observations that recombinant human UGT1A1 and UGT1A9, but not UGT1A4, catalysed the production of the major glucuronides of both genistein and daidzein in vitro. daidzein 181-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 18430559-7 2008 In view of a well-characterised functional polymorphism in UGT1A1, these observations may have implications for inter-individual variability in the potential health-beneficial effects of isoflavone consumption. Isoflavones 187-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 18797458-2 2008 We included 49 patients treated for metastatic colorectal cancer with a combination of 5-fluorouracil and irinotecan; a polymorphism in the UGT1A1 gene (TA repeat in the TATA box) and one in the CES2 gene promoter (830C>G) were studied as potential markers for SN-38 glucuronidation and irinotecan activation, respectively; and the potential activity of CYP3A4 was estimated from cortisol biotransformation into 6beta-hydroxycortisol. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 18797458-7 2008 The TATA box polymorphism of UGT1A1 was significantly associated with plasma bilirubin levels and behaved as a significant predictor for neutropoenia. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 18797458-10 2008 UGT1A1 genotyping plus cortisol 6beta-hydroxylation determination could help to determine the optimal dose of irinotecan. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18756540-4 2008 The UGT1A1 genotype significantly influenced average bilirubin levels for the common alleles: 6/6 genotype = 2.36 +/- 1.13 mg/dL, 6/7 genotype = 2.90 +/- 1.54 mg/dL, and 7/7 genotype = 4.24 +/- 2.11 mg/dL (P < 0.0001). Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 18756540-8 2008 These results validate previous smaller studies and confirm that the UGT1A1 promoter polymorphism exerts a powerful influence on bilirubin levels and the development of gallbladder disease in children with SCA. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 19004723-5 2008 Among the most conclusive examples one is that of the prediction of severe neutropenia induced by the irinotecan among patients homozygous for * 28 allele of UGT1A1 enzyme which conjugates SN38 active compound of irinotecan, the other one is the presence of a KRAS mutated allele in tumor cell to predict resistance to anti EGFR antibodies in the treatment of colorectal metastatic cancer. Irinotecan 213-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-141 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 18720361-1 2008 Pharmacogenetic research indicates a relationship between a polymorphism in the gene encoding uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and irinotecan inactivation, in that degradation of SN-38, the active metabolite of irinotecan, correlates inversely with the number of TA repeats in the TATA element of the UGT1A1 promoter region. Irinotecan 235-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 325-331 18720361-2 2008 Individuals who are homozygous for the UGT1A1*28 allele (7 repeats) may exhibit reduced degradation of SN-38 and increased probability of severe toxicities. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 18720361-5 2008 Irinotecan labeling recommends testing for the UGT1A1*28 allele and reducing irinotecan dosing in patients who are positive to reduce the likelihood of dose-limiting neutropenia only, but not diarrhea. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 18675828-6 2008 Overall, UGT1A1-53 and -3156 genotypes modified the association between dietary benzo(a)pyrene (BaP) and colon cancer (P for interaction=0.02 and 0.03, respectively). benzo(a) 80-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18653743-5 2008 UGT1A1 and UGT1A9 were the major enzymes responsible for the formation of the most abundant conjugate, isoliquiritigenin 4"-O-glucuronide (MG5), with Km values of 4.30+/-0.47 and 3.15+/-0.24 microM, respectively. isoliquiritigenin 4"-o-glucuronide 103-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18653743-5 2008 UGT1A1 and UGT1A9 were the major enzymes responsible for the formation of the most abundant conjugate, isoliquiritigenin 4"-O-glucuronide (MG5), with Km values of 4.30+/-0.47 and 3.15+/-0.24 microM, respectively. 3t83 139-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 19014835-3 2008 The protease inhibitor atazanavir is a potent inhibitor of UGT1A1. Atazanavir Sulfate 23-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 18653743-6 2008 UGT1A1 and UGT1A10 converted isoliquiritigenin to the next most abundant phase 2 metabolite, isoliquiritigenin 2"-O-glucuronide (MG4), with Km values of 2.98+/-0.8 and 25.8+/-1.3 microM, respectively. isoliquiritigenin 29-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18653743-6 2008 UGT1A1 and UGT1A10 converted isoliquiritigenin to the next most abundant phase 2 metabolite, isoliquiritigenin 2"-O-glucuronide (MG4), with Km values of 2.98+/-0.8 and 25.8+/-1.3 microM, respectively. isoliquiritigenin 2"-o-glucuronide 93-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18653743-6 2008 UGT1A1 and UGT1A10 converted isoliquiritigenin to the next most abundant phase 2 metabolite, isoliquiritigenin 2"-O-glucuronide (MG4), with Km values of 2.98+/-0.8 and 25.8+/-1.3 microM, respectively. mg4 129-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18675828-6 2008 Overall, UGT1A1-53 and -3156 genotypes modified the association between dietary benzo(a)pyrene (BaP) and colon cancer (P for interaction=0.02 and 0.03, respectively). pyrene 88-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18675828-6 2008 Overall, UGT1A1-53 and -3156 genotypes modified the association between dietary benzo(a)pyrene (BaP) and colon cancer (P for interaction=0.02 and 0.03, respectively). Benzo(a)pyrene 96-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18675828-7 2008 The strongest association was observed for those with <7.7 ng/day BaP exposure and the low activity genotypes, for both UGT1A1 28/28 (OR=1.8, 95% CI=1.1-2.9) and -3156AA (OR=1.7, 95% CI=1.0-3.0), compared to >or=7.7 ng/day and combined high/intermediate genotypes. Benzo(a)pyrene 69-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 18674515-13 2008 Kinetic analyses revealed that the main contributors to losartan glucuronidation in HLM are UGT1A1 and UGT2B7. Losartan 56-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 126-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18418374-7 2008 Polymorphisms in genes UGT1A1, UGT1A7, and UGT1A9 had strong associations with a component corresponding to the irinotecan-to-SN-38 pathway and SN-38 recirculation and to a component relating to SN-38-to-SN-38G conversion and elimination of SN-38G. Irinotecan 144-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18594531-1 2008 The aim of the study was to investigate the associations between UGT1A1(*)28 genotype and (1) response rates, (2) febrile neutropenia and (3) dose intensity in patients with metastatic colorectal cancer treated with irinotecan. Irinotecan 216-226 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 18781851-9 2008 Our results also demonstrate that possessing the *60/*60 plus *28/*28 diplotype in the UGT1A1 gene is a determinant of relatively higher bilirubin values amongst hyperbilirubinemic patients. Bilirubin 137-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 18566038-12 2008 Preliminary in vitro phenotyping studies indicated that glucuronide formation is primarily catalyzed by UGT1A1, 1A3, and 1A9. Glucuronides 56-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 17530442-0 2008 Induction of human UGT1A1 by a complex of dexamethasone-GR dependent on proximal site and independent of PBREM. Dexamethasone 42-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 17530442-1 2008 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays a key role to conjugate bilirubin and prevent jaundice. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 17530442-1 2008 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays a key role to conjugate bilirubin and prevent jaundice. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 17530442-4 2008 In this report, we investigated the influence of PBREM on the induction of the UGT1A1 reporter gene by GR and PXR with dexamethasone (DEX). Dexamethasone 119-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17530442-4 2008 In this report, we investigated the influence of PBREM on the induction of the UGT1A1 reporter gene by GR and PXR with dexamethasone (DEX). Dexamethasone 134-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17530442-10 2008 This supports that hPXR induced UGT1A1 through PBREM by DEX. Dexamethasone 56-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 17530442-11 2008 These results showed that PBREM has no relation with the induction by DEX-GR but the proximal site of UGT1A1 may function in stimulation by DEX-GR. dex-gr 140-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 18594531-2 2008 UGT1A1(*)28 genotype was determined in 218 patients receiving irinotecan (either first-line therapy with capecitabine or second-line as monotherapy) for metastatic colorectal cancer. Irinotecan 62-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18594531-2 2008 UGT1A1(*)28 genotype was determined in 218 patients receiving irinotecan (either first-line therapy with capecitabine or second-line as monotherapy) for metastatic colorectal cancer. Capecitabine 105-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18503403-5 2008 By performing uridine diphosphate glucuronosyltransferase (UGT) 1A1 genetic testing, some studies have been able to predict which patients receiving irinotecan will experience the toxicity. Irinotecan 149-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-67 18362158-3 2008 In this investigation we characterized the effects of albumin on the kinetics of 4-methylumbelliferone (4MU) glucuronidation by UDP-glucuronosyltransferase (UGT) 1A1, 1A6, and 1A9, and propofol (PRO) glucuronidation by UGT1A9 and HLM. Hymecromone 81-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-165 18362158-3 2008 In this investigation we characterized the effects of albumin on the kinetics of 4-methylumbelliferone (4MU) glucuronidation by UDP-glucuronosyltransferase (UGT) 1A1, 1A6, and 1A9, and propofol (PRO) glucuronidation by UGT1A9 and HLM. Hymecromone 104-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-165 18181169-4 2008 Independently, DHA and vitamin E resulted to affect only slightly UDP-glucuronosyltransferase 1A1 (UGT1A1) mRNA expression. Docosahexaenoic Acids 15-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-97 18181169-4 2008 Independently, DHA and vitamin E resulted to affect only slightly UDP-glucuronosyltransferase 1A1 (UGT1A1) mRNA expression. Docosahexaenoic Acids 15-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 18181169-4 2008 Independently, DHA and vitamin E resulted to affect only slightly UDP-glucuronosyltransferase 1A1 (UGT1A1) mRNA expression. Vitamin E 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-97 18181169-4 2008 Independently, DHA and vitamin E resulted to affect only slightly UDP-glucuronosyltransferase 1A1 (UGT1A1) mRNA expression. Vitamin E 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Bilirubin 191-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Irinotecan 236-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18172616-1 2008 Human UDP-glucuronosyltransferase (UGT)1A1 is a critical enzyme responsible for detoxification and metabolism of endogenous and exogenous lipophilic compounds, such as potentially neurotoxic bilirubin and the anticancer drug irinotecan SN-38, via conjugation with glucuronic acid. Glucuronic Acid 264-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-42 18172616-2 2008 A 290-bp distal enhancer module, phenobarbital-responsive enhancer module of UGT1A1 (gtPBREM), fully accounts for constitutive androstane receptor (CAR)-, pregnane X receptor (PXR)-, glucocorticoid receptor (GR)-, and aryl hydrocarbon receptor (AhR)-mediated activation of the UGT1A1 gene. Phenobarbital 33-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 17700594-1 2008 Genetic polymorphisms in hepatically expressed UGT1A1 and UGT1A9 contribute to the interindividual variability i-n irinotecan disposition and toxicity. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 17700594-5 2008 UGT1A1-1A9 diplotype analysis showed that patients harbouring the H1/H2 (TG6GT(10)T/GG6GT(9)C) diplotype had 2.4-fold lower systemic exposure to SN-38 glucuronide (SN-38G) compared with patients harbouring the H1/H5 (TG6GT(10)T/GG6GT(10)C) diplotype (P=0.025). 7-ethyl-10-hydroxycamptothecin glucuronide 145-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17700594-5 2008 UGT1A1-1A9 diplotype analysis showed that patients harbouring the H1/H2 (TG6GT(10)T/GG6GT(9)C) diplotype had 2.4-fold lower systemic exposure to SN-38 glucuronide (SN-38G) compared with patients harbouring the H1/H5 (TG6GT(10)T/GG6GT(10)C) diplotype (P=0.025). Irinotecan 145-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18633245-0 2008 [Role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction]. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 18633245-0 2008 [Role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction]. Irinotecan 36-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 76-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-3 2008 Irinotecan is metabolized by carboxylesterase to form an active metabolite, 7-ethyl-10-hydroxycamptothecin(SN-38), which in turn is subsequently conjugated by UGT-glucuronosyltransferase 1A1(UGT1A1)to yield an inactive form, SN-38 glucuronide(SN-38 G). Irinotecan 107-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 18633245-4 2008 The UGT1A1 gene polymorphisms contribute to the individual variation in adverse events among patients administered irinotecan. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 18633245-7 2008 Recently, it has been demonstrated that genetic variants of UGT1A1*6 in addition to UGT1A1*28 are associated with the occurrence of adverse events in irinotecan chemotherapy in Asians. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 18633245-7 2008 Recently, it has been demonstrated that genetic variants of UGT1A1*6 in addition to UGT1A1*28 are associated with the occurrence of adverse events in irinotecan chemotherapy in Asians. Irinotecan 150-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 18633245-8 2008 This review summarizes recent studies to outline the role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction in Japanese cancer patients. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 18633245-8 2008 This review summarizes recent studies to outline the role of UGT1A1*28 and UGT1A1*6 for irinotecan-induced adverse drug reaction in Japanese cancer patients. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 18558634-5 2008 RESULTS: Genetic polymorphisms of the UGT1A1 promoter, specifically the T-3279G phenobarbital responsive enhancer module and the (thymidine-adenine)(7) dinucleotide repeat TATAA box variants, were common. Phenobarbital 80-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 18362158-0 2008 The "albumin effect" and drug glucuronidation: bovine serum albumin and fatty acid-free human serum albumin enhance the glucuronidation of UDP-glucuronosyltransferase (UGT) 1A9 substrates but not UGT1A1 and UGT1A6 activities. Fatty Acids 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 18633225-1 2008 We encountered a 64-year-old UGT1A1*28 heterotype patient who received CPT-11 and CDDP chemotherapy for Stage III A small-cell lung-cancer and developed grade 4 neutropenia as assessed by CTCAE v3.0 in Pharmaceutical Management. Irinotecan 71-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 18633225-1 2008 We encountered a 64-year-old UGT1A1*28 heterotype patient who received CPT-11 and CDDP chemotherapy for Stage III A small-cell lung-cancer and developed grade 4 neutropenia as assessed by CTCAE v3.0 in Pharmaceutical Management. cddp 82-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 18172616-2 2008 A 290-bp distal enhancer module, phenobarbital-responsive enhancer module of UGT1A1 (gtPBREM), fully accounts for constitutive androstane receptor (CAR)-, pregnane X receptor (PXR)-, glucocorticoid receptor (GR)-, and aryl hydrocarbon receptor (AhR)-mediated activation of the UGT1A1 gene. Phenobarbital 33-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 277-283 18172616-6 2008 Thus, UGT1A1 gene promoter variations, including the TA repeat polymorphism and T-3279G gtPBREM, have important clinical implications. gtpbrem 88-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 18466101-0 2008 Cost-effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Irinotecan 86-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 18541046-12 2008 We also estimates the potential effects of genetic variations: the AUC and Cmax of SN-38 may increase to 208% and 165% respectively with the genetic variation UGT1A1*28/*28 which reduces the expression of UGT1A1 down to 30%. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 18541046-12 2008 We also estimates the potential effects of genetic variations: the AUC and Cmax of SN-38 may increase to 208% and 165% respectively with the genetic variation UGT1A1*28/*28 which reduces the expression of UGT1A1 down to 30%. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 18343383-0 2008 Gilbert syndrome, UGT1A1*28 allele, and cardiovascular disease risk: possible protective effects and therapeutic applications of bilirubin. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 18343383-5 2008 HO and biliverdin reductase control the formation of bilirubin, whereas UGT1A1 controls bilirubin conjugation and clearance. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 18300238-0 2008 UGT1A1*28 polymorphism predicts irinotecan-induced severe toxicities without affecting treatment outcome and survival in patients with metastatic colorectal carcinoma. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-78 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 18300238-1 2008 BACKGROUND: It is known that the uridine-diphosphoglucuronosyl transferase 1A1 (UGT1A1)*28 polymorphism reduces UGT1A1 enzyme activity, which may lead to severe toxicities in patients who receive irinotecan. Irinotecan 196-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 18300238-11 2008 CONCLUSIONS: The current data suggested that the UGT1A1*28 polymorphism may be a key determinant for predicting irinotecan-induced severe toxicities without affecting treatment outcome for patients with metastatic CRC. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 18375480-0 2008 Association between the UGT1A1 TA-repeat polymorphism and bilirubin concentration in patients with intermittent claudication: results from the CAVASIC study. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 18375480-2 2008 Low plasma concentrations of bilirubin are reportedly associated with the development of coronary and cerebrovascular disease, and bilirubin concentrations are strongly correlated with the enzyme activity of the hepatic uridine diphosphate glucuronosyltransferase (UGT1A1). Bilirubin 131-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 265-271 18375480-4 2008 In a case-control study, we investigated the association between the UGT1A1 polymorphism, bilirubin concentration, and intermittent claudication. Bilirubin 90-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 19372981-6 2008 Some other important advances in our knowledge have also been made, such as the association of TA repeats in the UGT1A1 regulatory region (UGT1A1 28) with atazanavir-related hyperbilirubinaemia and the association of ABCC2 and ABCC4 single nucleotide polymorphisms with tenofovir-associated renal toxicity. Atazanavir Sulfate 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 19372981-6 2008 Some other important advances in our knowledge have also been made, such as the association of TA repeats in the UGT1A1 regulatory region (UGT1A1 28) with atazanavir-related hyperbilirubinaemia and the association of ABCC2 and ABCC4 single nucleotide polymorphisms with tenofovir-associated renal toxicity. Atazanavir Sulfate 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 19372981-6 2008 Some other important advances in our knowledge have also been made, such as the association of TA repeats in the UGT1A1 regulatory region (UGT1A1 28) with atazanavir-related hyperbilirubinaemia and the association of ABCC2 and ABCC4 single nucleotide polymorphisms with tenofovir-associated renal toxicity. Tenofovir 270-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 19372981-6 2008 Some other important advances in our knowledge have also been made, such as the association of TA repeats in the UGT1A1 regulatory region (UGT1A1 28) with atazanavir-related hyperbilirubinaemia and the association of ABCC2 and ABCC4 single nucleotide polymorphisms with tenofovir-associated renal toxicity. Tenofovir 270-279 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 18652401-4 2008 Polymorphisms of UGT 1A1 are considered as potential indicators of a risk of toxicity treatment by irinotecan. Irinotecan 99-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-24 18437170-4 2008 In addition, the Food and Drug Administration of the United States now requires a "black box" warning on the packaging of irinotecan for evaluation of germline polymorphism in UGT1A1, the gene mutated in Gilbert"s syndrome, for potential reduction of drug dosage in patients with the UGT1A1*28 polymorphism. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 18437170-4 2008 In addition, the Food and Drug Administration of the United States now requires a "black box" warning on the packaging of irinotecan for evaluation of germline polymorphism in UGT1A1, the gene mutated in Gilbert"s syndrome, for potential reduction of drug dosage in patients with the UGT1A1*28 polymorphism. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 284-290 18466101-1 2008 AIM: The aim of the present study was to evaluate the cost-effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 18466101-5 2008 RESULTS & CONCLUSION: Genotyping in combination with a subsequent reduction of initial irinotecan dose for UGT1A1 7/7 genotype patients was cost-saving for the population of African and Caucasian origin. Adenosine Monophosphate 9-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 18466101-5 2008 RESULTS & CONCLUSION: Genotyping in combination with a subsequent reduction of initial irinotecan dose for UGT1A1 7/7 genotype patients was cost-saving for the population of African and Caucasian origin. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 18466101-8 2008 Finally, the application of a 3-weekly high-dose treatment regimen with a 20% reduced dosage compared with the low-dose weekly irinotecan regimen in patients with UGT1A1 7/7 genotype was less expensive and is more convenient for the patient. Irinotecan 127-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 18379174-2 2008 Irinotecan, an anticancer drug, is one of the models for personalized medicine, and a number of clinical studies have revealed significant associations between UGT1A1(*)28 and irinotecan toxicity. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 18328680-6 2008 PB induced CYP3A4, 3A5, 2B6 and 2A6, UGT1A1 and all transporters. Phenobarbital 0-2 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 18021224-3 2008 uridine 5"-diphosphate-glucuronosyltransferase 1A1 (UGT1A1) glucuronidates bilirubin for solubilization in the ER. Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-50 18021224-3 2008 uridine 5"-diphosphate-glucuronosyltransferase 1A1 (UGT1A1) glucuronidates bilirubin for solubilization in the ER. Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 18021224-6 2008 CONCLUSION: These results strongly suggest that bilirubin is directly delivered to UGT1A1 from ligandin for glucuronidation. Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. naringenin 8-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 18248513-6 2008 Both of naringenin and elacridar separately enhanced the sensitivity for CPT-11 and SN-38 in KYN-2 cells abundantly expressing BCRP, CYP3A4/5 and UGT1A1, but not in KYN-1 cells expressing lower levels. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 18098330-7 2008 It also aimed at relating glucuronidation of riluzole to differential UGT1A1*28 genotypes. Riluzole 45-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 17713471-5 2008 The LPV/RTV treatment also inhibited the formation of SN38 glucuronide (SN38G), as shown by the 36% decrease in the SN38G/SN38 AUCs ratio (5.9+/-1.6 vs 9.2+/-2.6, P=0.002) consistent with UGT1A1 inhibition by LPV/RTV. 7-ethyl-10-hydroxycamptothecin glucuronide 54-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 17713471-5 2008 The LPV/RTV treatment also inhibited the formation of SN38 glucuronide (SN38G), as shown by the 36% decrease in the SN38G/SN38 AUCs ratio (5.9+/-1.6 vs 9.2+/-2.6, P=0.002) consistent with UGT1A1 inhibition by LPV/RTV. Lopinavir 4-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 17713471-5 2008 The LPV/RTV treatment also inhibited the formation of SN38 glucuronide (SN38G), as shown by the 36% decrease in the SN38G/SN38 AUCs ratio (5.9+/-1.6 vs 9.2+/-2.6, P=0.002) consistent with UGT1A1 inhibition by LPV/RTV. Ritonavir 8-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 18187562-0 2008 Contribution of UDP-glucuronosyltransferase 1A1 and 1A8 to morphine-6-glucuronidation and its kinetic properties. Morphine 59-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-55 18187562-5 2008 Not only UGT2B7, which is well known to catalyze morphine-6-glucuronidation, but also UGT1A1 and 1A8 effectively catalyzed morphine-6-glucuronidation at relatively low morphine concentrations (<100 muM). Morphine 49-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 18187562-5 2008 Not only UGT2B7, which is well known to catalyze morphine-6-glucuronidation, but also UGT1A1 and 1A8 effectively catalyzed morphine-6-glucuronidation at relatively low morphine concentrations (<100 muM). Morphine 123-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 18187562-5 2008 Not only UGT2B7, which is well known to catalyze morphine-6-glucuronidation, but also UGT1A1 and 1A8 effectively catalyzed morphine-6-glucuronidation at relatively low morphine concentrations (<100 muM). Morphine 123-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 18187562-11 2008 The results strongly suggest that UGT1A1 and UGT1A8 are isozymes involved in morphine-6-glucuronidation in vivo, as is UGT2B7 in humans. Morphine 77-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 18467239-0 2008 UGT1A1*28 and other UGT1A polymorphisms as determinants of irinotecan toxicity. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18379174-2 2008 Irinotecan, an anticancer drug, is one of the models for personalized medicine, and a number of clinical studies have revealed significant associations between UGT1A1(*)28 and irinotecan toxicity. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 18379174-6 2008 This mini-review outlines our recent studies on irinotecan pharmacogenetics and discusses the clinical significance of UGT1A1(*)6 and (*)28 markers for personalized irinotecan therapy in Japanese cancer patients. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 18180294-1 2008 The 9 UDP-glucuronosyltranferases (UGTs) encoded by the UGT1 locus in humans are key enzymes in the metabolism of most drugs as well as endogenous substances such as bile acids, fatty acids, steroids, hormones, neurotransmitters, and bilirubin. Bile Acids and Salts 166-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-60 18180294-1 2008 The 9 UDP-glucuronosyltranferases (UGTs) encoded by the UGT1 locus in humans are key enzymes in the metabolism of most drugs as well as endogenous substances such as bile acids, fatty acids, steroids, hormones, neurotransmitters, and bilirubin. Fatty Acids 178-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-60 18180294-1 2008 The 9 UDP-glucuronosyltranferases (UGTs) encoded by the UGT1 locus in humans are key enzymes in the metabolism of most drugs as well as endogenous substances such as bile acids, fatty acids, steroids, hormones, neurotransmitters, and bilirubin. Steroids 191-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-60 18180294-1 2008 The 9 UDP-glucuronosyltranferases (UGTs) encoded by the UGT1 locus in humans are key enzymes in the metabolism of most drugs as well as endogenous substances such as bile acids, fatty acids, steroids, hormones, neurotransmitters, and bilirubin. Bilirubin 234-243 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-60 18180294-4 2008 Because UGT1A1 in humans is responsible for 100% of the conjugated bilirubin, it followed that newborn Ugt1(-/-) mice developed serum levels of unconjugated bilirubin that were 40-60 times higher than Ugt1(+/-) or wild-type mice. Bilirubin 67-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 18180294-4 2008 Because UGT1A1 in humans is responsible for 100% of the conjugated bilirubin, it followed that newborn Ugt1(-/-) mice developed serum levels of unconjugated bilirubin that were 40-60 times higher than Ugt1(+/-) or wild-type mice. Bilirubin 157-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-107 18180294-5 2008 The result of extreme unconjugated bilirubin in Ugt1(-/-) mice, comparable to the induced levels noted in patients with Crigler-Najjar type 1 disease, is fatal in neonatal Ugt1(-/-) mice within 2 weeks following birth. Bilirubin 35-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-176 18349289-7 2008 CONCLUSIONS: Irinotecan toxicity is more likely in patients with Gilbert"s syndrome carrying the UGT1A1*28 allele combined with reduced function UGT1A7 N129K/R131K and UGT1A7-57T/G SNP. Irinotecan 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 18082937-0 2008 Importance of UDP-glucuronosyltransferase 1A1*6 for irinotecan toxicities in Japanese cancer patients. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-45 18082937-1 2008 Recent pharmacogenetic studies on irinotecan have revealed the impact of UDP glucuronosyltransferase (UGT) 1A1*28 on severe irinotecan toxicities. Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-110 18082937-1 2008 Recent pharmacogenetic studies on irinotecan have revealed the impact of UDP glucuronosyltransferase (UGT) 1A1*28 on severe irinotecan toxicities. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-110 18082937-2 2008 Although the clinical role of UGT1A1*6, which is specifically detected in East Asian patients, in irinotecan toxicities is suggested, clear evidence remains limited. Irinotecan 98-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 18082937-3 2008 To examine the impact of *6, the association of UGT1A1 genotypes with severe irinotecan toxicities was retrospectively investigated in Japanese cancer patients. Irinotecan 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 18349289-1 2008 BACKGROUND: Gilbert"s syndrome is characterized by a functional promoter single nucleotide polymorphism (SNP) of the UDP-glucuronosyltransferase (UGT) 1A1 gene and represents a pharmacogenetic risk factor for irinotecan toxicity, but study data remain controversial. Irinotecan 209-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-154 18349289-8 2008 Based on the ability of UGT1A7 to metabolize and eliminate the active irinotecan metabolite 7-ethyl-10-hydroxycamptothecin, the UGT1A1/UGT1A7 SNP combination haplotype appears to be a superior risk predictor than Gilbert"s syndrome alone. Irinotecan 92-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Steroids 187-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-62 19255595-7 2008 Continual dietary exposure to chrysin and quercetin, found in fruits and vegetables, induces UGT1A1 and may reduce exposure to hydroxylated PCBs through increased glucuronidation. chrysin 30-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 19255595-7 2008 Continual dietary exposure to chrysin and quercetin, found in fruits and vegetables, induces UGT1A1 and may reduce exposure to hydroxylated PCBs through increased glucuronidation. Quercetin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 37-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. 7-ethyl-10-hydroxycamptothecin glucuronide 105-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-1 2008 CPT-11 is activated by hydrolysis to SN-38 that is primarily inactivated through biotrans-formation into SN-38 glucuronide ( SN-38 G) by UGT1As. Irinotecan 105-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-141 18281753-3 2008 The toxicities of CPT-11 have been reported to correlate with the polymorphism of the number of TA repeats in UGT1A1 gene because of its gene transcriptional efficiency. Irinotecan 18-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 17998297-3 2008 In the present study, we found that UGT1A1-catalyzed estradiol 3-O-glucuronide formation and UGT1A4-catalyzed imipramine N-glucuronide formation in human liver microsomes were prominently decreased in the presence of 1-naphthol, but those by recombinant human UGT1A1 and UGT1A4, respectively, were not. estradiol 3-o-glucuronide 53-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 17998297-3 2008 In the present study, we found that UGT1A1-catalyzed estradiol 3-O-glucuronide formation and UGT1A4-catalyzed imipramine N-glucuronide formation in human liver microsomes were prominently decreased in the presence of 1-naphthol, but those by recombinant human UGT1A1 and UGT1A4, respectively, were not. imipramine N-glucuronide 110-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 260-266 17998297-3 2008 In the present study, we found that UGT1A1-catalyzed estradiol 3-O-glucuronide formation and UGT1A4-catalyzed imipramine N-glucuronide formation in human liver microsomes were prominently decreased in the presence of 1-naphthol, but those by recombinant human UGT1A1 and UGT1A4, respectively, were not. 1-naphthol 217-227 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 17998297-4 2008 Interestingly, when recombinant UGT1A6 was added in the reaction mixture, these activities by recombinant UGT1A1 and UGT1A4 were diminished in the presence of 1-naphthol. 1-naphthol 159-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 17998297-6 2008 We found that UDP strongly inhibited the UGT1A1 (K(i) = 7 microM) and UGT1A4 (K(i) = 47 microM) activities in a competitive manner for the 5"-diphosphoglucuronic acid binding. Uridine Diphosphate 14-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 17998297-6 2008 We found that UDP strongly inhibited the UGT1A1 (K(i) = 7 microM) and UGT1A4 (K(i) = 47 microM) activities in a competitive manner for the 5"-diphosphoglucuronic acid binding. 5"-diphosphoglucuronic acid 139-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 17998297-9 2008 Among them, 4 compounds (1-naphthol, 2-naphthol, 4-nitrophenol, and 4-methylumbelliferone) with high turnover rates (V(max)/K(m) value >200 mul/min/mg) showed more potent inhibition of the activity in human liver microsomes compared with that by the recombinant UGT1A1. 1-naphthol 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 265-271 17998297-9 2008 Among them, 4 compounds (1-naphthol, 2-naphthol, 4-nitrophenol, and 4-methylumbelliferone) with high turnover rates (V(max)/K(m) value >200 mul/min/mg) showed more potent inhibition of the activity in human liver microsomes compared with that by the recombinant UGT1A1. 2-naphthol 37-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 265-271 17998297-9 2008 Among them, 4 compounds (1-naphthol, 2-naphthol, 4-nitrophenol, and 4-methylumbelliferone) with high turnover rates (V(max)/K(m) value >200 mul/min/mg) showed more potent inhibition of the activity in human liver microsomes compared with that by the recombinant UGT1A1. 4-nitrophenol 49-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 265-271 17998297-9 2008 Among them, 4 compounds (1-naphthol, 2-naphthol, 4-nitrophenol, and 4-methylumbelliferone) with high turnover rates (V(max)/K(m) value >200 mul/min/mg) showed more potent inhibition of the activity in human liver microsomes compared with that by the recombinant UGT1A1. Hymecromone 68-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 265-271 17952380-3 2008 Unconjugated bilirubin is conjugated with glucuronic acid through the reaction of uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1). Bilirubin 13-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-133 17952380-3 2008 Unconjugated bilirubin is conjugated with glucuronic acid through the reaction of uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1). Bilirubin 13-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 17952380-3 2008 Unconjugated bilirubin is conjugated with glucuronic acid through the reaction of uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1). Glucuronic Acid 42-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-133 17952380-3 2008 Unconjugated bilirubin is conjugated with glucuronic acid through the reaction of uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1). Glucuronic Acid 42-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 17952380-10 2008 Although the concentration of bilirubin in the UGT1A1 variant was higher than the wild type for the patients in the CAD group, there was no significant difference in the polymorphism of UGT1A1 between the patients in the CAD group and the participants in the control group. Bilirubin 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Steroids 187-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-69 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Bile Acids and Salts 205-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-62 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Bile Acids and Salts 205-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-69 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Bilirubin 221-230 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-62 19003600-1 2008 The substrate spectrum of human UDP-glucuronosyltransferase 1A (UGT1A) proteins includes the glucuronidation of non-steroidal anti-inflammatory drugs, anticonvulsants, chemotherapeutics, steroid hormones, bile acids, and bilirubin. Bilirubin 221-230 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-69 18392554-2 2008 We investigated whether the (TA)n promoter polymorphism in the UDP-glucuronosyltransferase 1A1 gene (UGT1A1) may modify bilirubin metabolism, influencing bilirubinaemia, predisposition to cholelithiasis and subsequent cholecystectomy, in a group of 153 young SCD patients (mean age 12.0 +/- 9.0 years) predominantly of Bantu beta S haplotype. Bilirubin 120-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-94 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Rifampin 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Dexamethasone 5-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Omeprazole 14-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Omeprazole 102-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Rifampin 134-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18305373-4 2008 Rif, Dex, and Ome increased the mRNA level of UGT1A1 in both human and cynomolgus monkey hepatocytes, Ome more potently in humans and Rif and Ome more potently in monkeys. Omeprazole 102-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 18095922-5 2008 Raltegravir is not a potent inhibitor or inducer of cytochrome P450 3A4, and it is predominantly metabolized by glucuronidation, specifically by the enzyme UDP-glucuronosyltransferase 1A1. Raltegravir Potassium 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-187 18392554-2 2008 We investigated whether the (TA)n promoter polymorphism in the UDP-glucuronosyltransferase 1A1 gene (UGT1A1) may modify bilirubin metabolism, influencing bilirubinaemia, predisposition to cholelithiasis and subsequent cholecystectomy, in a group of 153 young SCD patients (mean age 12.0 +/- 9.0 years) predominantly of Bantu beta S haplotype. Bilirubin 120-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 18098064-0 2008 Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Progesterone 55-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-51 17921187-4 2008 HLMs, UGT1A1, 1A8, 1A9, and 1A10 showed glucuronidation activity toward 4"-, 6-, 7-, and/or 8-hydroxywarfarin with K(m) values ranging from 59 to 480 microM and V(max) values ranging from 0.03 to 0.78 microM/min/mg protein. 4"-, 6-, 7-, and/ 72-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 17921187-4 2008 HLMs, UGT1A1, 1A8, 1A9, and 1A10 showed glucuronidation activity toward 4"-, 6-, 7-, and/or 8-hydroxywarfarin with K(m) values ranging from 59 to 480 microM and V(max) values ranging from 0.03 to 0.78 microM/min/mg protein. 8-hydroxywarfarin 92-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 17921187-6 2008 Of the hepatic UGT isozymes tested, UGT1A9 exclusively metabolized 8-hydroxywarfarin, whereas UGT1A1 metabolized 6-, 7-, and 8-hydroxywarfarin. 6-, 7-, and 8-hydroxywarfarin 113-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 18043502-3 2008 (TA)7 promoter polymorphism in the gene encoding the bilirubin conjugating enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1) potentiates hyperbilirubinemia in G-6-PD deficient neonates. Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-113 18043502-3 2008 (TA)7 promoter polymorphism in the gene encoding the bilirubin conjugating enzyme UDP-glucuronosyltransferase 1A1 (UGT1A1) potentiates hyperbilirubinemia in G-6-PD deficient neonates. Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 18098064-0 2008 Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Labetalol 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-51 18098064-3 2008 Labetalol glucuronidation, which is the major elimination pathway of labetalol, was characterized by screening six recombinant human UGTs (UGT1A1, 1A4, 1A6, 1A9, 2B4, and 2B7) for their capacity to catalyse labetalol glucuronidation. Labetalol 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 18098064-6 2008 UGT1A1 and UGT2B7 were identified as the major UGT enzymes producing labetalol glucuronides (trace amount of glucuronide conjugate was formed by UGT1A9). labetalol glucuronides 69-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18098064-6 2008 UGT1A1 and UGT2B7 were identified as the major UGT enzymes producing labetalol glucuronides (trace amount of glucuronide conjugate was formed by UGT1A9). Glucuronides 79-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 18098064-7 2008 The activities of the UGT1A1 promoter containing PXR response elements were enhanced by progesterone, but not by oestrogens, indicating PXR-mediated induction of UGT1A1 promoter activity by progesterone. Progesterone 88-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 18098064-7 2008 The activities of the UGT1A1 promoter containing PXR response elements were enhanced by progesterone, but not by oestrogens, indicating PXR-mediated induction of UGT1A1 promoter activity by progesterone. Progesterone 88-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 18098064-7 2008 The activities of the UGT1A1 promoter containing PXR response elements were enhanced by progesterone, but not by oestrogens, indicating PXR-mediated induction of UGT1A1 promoter activity by progesterone. Progesterone 190-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 18098064-7 2008 The activities of the UGT1A1 promoter containing PXR response elements were enhanced by progesterone, but not by oestrogens, indicating PXR-mediated induction of UGT1A1 promoter activity by progesterone. Progesterone 190-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 18098064-11 2008 The results suggest a potential role for progesterone in regulating labetalol elimination by modulating the expression of UGT1A1, leading to enhanced drug metabolism during pregnancy. Labetalol 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 17761779-7 2007 Mefenamic acid, a UGT1A9 inhibitor, decreased FYX-051 N(1)- and N(2)-glucuronosyltransferase activities, whereas bilirubin, a UGT1A1 inhibitor, did not affect these activities. Bilirubin 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 126-132 17956868-10 2007 Furthermore, mutation of His-39 residue of UGT1A1 (His-40 in UGT1A4) to proline led to loss of glucuronidation of phenols but not of estrogens. Histidine 25-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17956868-10 2007 Furthermore, mutation of His-39 residue of UGT1A1 (His-40 in UGT1A4) to proline led to loss of glucuronidation of phenols but not of estrogens. Histidine 51-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17956868-10 2007 Furthermore, mutation of His-39 residue of UGT1A1 (His-40 in UGT1A4) to proline led to loss of glucuronidation of phenols but not of estrogens. Proline 72-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17956868-10 2007 Furthermore, mutation of His-39 residue of UGT1A1 (His-40 in UGT1A4) to proline led to loss of glucuronidation of phenols but not of estrogens. Phenols 114-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17981384-6 2007 Compared with control cells (MCF-7(VEC)), reduced cell proliferation was seen in cells expressing UGT1A1 (MCF-7(UGT1A1)) under estradiol treatment. Estradiol 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17981384-6 2007 Compared with control cells (MCF-7(VEC)), reduced cell proliferation was seen in cells expressing UGT1A1 (MCF-7(UGT1A1)) under estradiol treatment. Estradiol 127-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 17981384-8 2007 Over-expressing UGT1A1 reduced the binding of DMBA to DNA, and increased MCF-7(UGT1A1) intact cells under DMBA treatment was verified by comet assay. 9,10-Dimethyl-1,2-benzanthracene 46-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 17981384-8 2007 Over-expressing UGT1A1 reduced the binding of DMBA to DNA, and increased MCF-7(UGT1A1) intact cells under DMBA treatment was verified by comet assay. 9,10-Dimethyl-1,2-benzanthracene 106-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 17981384-8 2007 Over-expressing UGT1A1 reduced the binding of DMBA to DNA, and increased MCF-7(UGT1A1) intact cells under DMBA treatment was verified by comet assay. 9,10-Dimethyl-1,2-benzanthracene 106-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17875670-5 2007 All the UGTs examined catalyzed the formation of T4 phenolic glucuronide except UGT1A4; the highest activity was detected with UGT1A3, UGT1A8, and UGT1A10, followed by UGT1A1 and UGT2B4. Glucuronides 61-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 147-153 18004206-2 2007 This work compared the effects of (a) the individual UGT1A1 mutations on the glucuronidation kinetics bilirubin, beta-estradiol, 4-methylumbelliferone (4MU) and 1-naphthol (1NP), and (b) the Y486 mutation, which occurs in the conserved carboxyl terminal domain of UGT1A enzymes, on 4MU, 1NP and naproxen glucuronidation by UGT1A3, UGT1A6 and UGT1A10. Bilirubin 102-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 17875670-6 2007 Formation of T3 phenolic glucuronide was observed in the order of UGT1A8 > UGT1A10 > UGT1A3 > UGT1A1; trace activity was observed with UGT1A6 and UGT1A9. t3 phenolic glucuronide 13-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 17898154-1 2007 The UGT1A1*28 polymorphism is known to correlate with altered clearance of bilirubin (Gilbert syndrome) and drugs such as 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11). Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17898154-1 2007 The UGT1A1*28 polymorphism is known to correlate with altered clearance of bilirubin (Gilbert syndrome) and drugs such as 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11). Irinotecan 122-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17898154-1 2007 The UGT1A1*28 polymorphism is known to correlate with altered clearance of bilirubin (Gilbert syndrome) and drugs such as 7-ethyl-10-[4-(1-piperidino)-1-piperidino] carbonyloxy camptothecin (CPT-11). Irinotecan 191-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17898154-5 2007 Bilirubin, estradiol (3-OH), ethinyl estradiol (3-OH), and 7-ethyl-10-hydroxycamptothecin (SN-38) were found to show significantly lower rates of metabolism in the UGT1A1*28/*28 microsomes with no change in K(m) values. Irinotecan 59-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 17898154-8 2007 Interestingly, muraglitazar (UGT1A3 substrate) showed an inverse correlation with glucuronidation of UGT1A1 substrates. muraglitazar 15-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 18004206-2 2007 This work compared the effects of (a) the individual UGT1A1 mutations on the glucuronidation kinetics bilirubin, beta-estradiol, 4-methylumbelliferone (4MU) and 1-naphthol (1NP), and (b) the Y486 mutation, which occurs in the conserved carboxyl terminal domain of UGT1A enzymes, on 4MU, 1NP and naproxen glucuronidation by UGT1A3, UGT1A6 and UGT1A10. Estradiol 113-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 18004206-2 2007 This work compared the effects of (a) the individual UGT1A1 mutations on the glucuronidation kinetics bilirubin, beta-estradiol, 4-methylumbelliferone (4MU) and 1-naphthol (1NP), and (b) the Y486 mutation, which occurs in the conserved carboxyl terminal domain of UGT1A enzymes, on 4MU, 1NP and naproxen glucuronidation by UGT1A3, UGT1A6 and UGT1A10. Hymecromone 129-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 18004206-5 2007 RESULTS: Compared with wild-type UGT1A1, in-vitro clearances for bilirubin, beta-estradiol, 4MU and 1NP glucuronidation by UGT1A1*6 and UGT1A1*27 were reduced by 34-74%, most commonly as a result of a reduction in Vmax. Bilirubin 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 18004206-5 2007 RESULTS: Compared with wild-type UGT1A1, in-vitro clearances for bilirubin, beta-estradiol, 4MU and 1NP glucuronidation by UGT1A1*6 and UGT1A1*27 were reduced by 34-74%, most commonly as a result of a reduction in Vmax. Bilirubin 65-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 18004206-5 2007 RESULTS: Compared with wild-type UGT1A1, in-vitro clearances for bilirubin, beta-estradiol, 4MU and 1NP glucuronidation by UGT1A1*6 and UGT1A1*27 were reduced by 34-74%, most commonly as a result of a reduction in Vmax. Estradiol 76-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 18004206-5 2007 RESULTS: Compared with wild-type UGT1A1, in-vitro clearances for bilirubin, beta-estradiol, 4MU and 1NP glucuronidation by UGT1A1*6 and UGT1A1*27 were reduced by 34-74%, most commonly as a result of a reduction in Vmax. Estradiol 76-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 17927138-5 2007 The results also indicated that well-expressed UGT isoforms in the Caco-2 cells, UGT1A1, UGT1A3, UGT1A6, and UGT2B7, were capable of metabolizing apigenin faster than genistein and that UGT1A6 silencing did not substantially increase the level of expression of genistein-metabolizing UGT isoforms. Apigenin 146-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 17978490-0 2007 Stereoselective metabolism of racemic carvedilol by UGT1A1 and UGT2B7, and effects of mutation of these enzymes on glucuronidation activity. Carvedilol 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 17978490-1 2007 Carvedilol, an alpha- and beta-adrenergic blocking drug, is mainly metabolized by CYP2D6, UGT1A1, UGT2B4 and UGT2B7. Carvedilol 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 17978490-6 2007 The aim of this study was to clarify stereoselective metabolism of carvedilol by UGT1A1 and UGT2B7 and to determine by using a recombinant enzyme-introduced mutation whether genetic mutation in UGT1A1 and UGT2B7 causes reduction in metabolic activity for carvedilol. Carvedilol 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 17978490-6 2007 The aim of this study was to clarify stereoselective metabolism of carvedilol by UGT1A1 and UGT2B7 and to determine by using a recombinant enzyme-introduced mutation whether genetic mutation in UGT1A1 and UGT2B7 causes reduction in metabolic activity for carvedilol. Carvedilol 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 17978490-6 2007 The aim of this study was to clarify stereoselective metabolism of carvedilol by UGT1A1 and UGT2B7 and to determine by using a recombinant enzyme-introduced mutation whether genetic mutation in UGT1A1 and UGT2B7 causes reduction in metabolic activity for carvedilol. Carvedilol 255-265 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 17978490-7 2007 A glucuronidation assay using human liver microsomes and recombinant UGT1A1 and UGT2B7 expressed in HeLa cells demonstrated that UGT1A1 prefers metabolizing R-carvedilol to S-carvedilol. Carvedilol 157-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 17978490-7 2007 A glucuronidation assay using human liver microsomes and recombinant UGT1A1 and UGT2B7 expressed in HeLa cells demonstrated that UGT1A1 prefers metabolizing R-carvedilol to S-carvedilol. Carvedilol 157-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 17978490-7 2007 A glucuronidation assay using human liver microsomes and recombinant UGT1A1 and UGT2B7 expressed in HeLa cells demonstrated that UGT1A1 prefers metabolizing R-carvedilol to S-carvedilol. Carvedilol 173-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 17611564-5 2007 As nilotinib is not known to be glucuronidated by UGT1A1, the combined impact of inhibition of UGT1A1 activity by nilotinib and genetic polymorphism is the most likely cause of the increased rate of hyperbilirubinemia. nilotinib 3-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 17611564-5 2007 As nilotinib is not known to be glucuronidated by UGT1A1, the combined impact of inhibition of UGT1A1 activity by nilotinib and genetic polymorphism is the most likely cause of the increased rate of hyperbilirubinemia. nilotinib 114-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 17888052-1 2007 AIM: To investigate bilirubin UDP-glucuronosyltransferase (UGT1A1) gene allele in healthy Chinese neonates, their cord bilirubin level and the subsequent hyperbilirubinemia to determine relationships among them. Bilirubin 20-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 17888052-1 2007 AIM: To investigate bilirubin UDP-glucuronosyltransferase (UGT1A1) gene allele in healthy Chinese neonates, their cord bilirubin level and the subsequent hyperbilirubinemia to determine relationships among them. Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 17670842-6 2007 Evaluation of 12 recombinant human UGT isozymes shows gemfibrozil glucuronidation activity in UGT1A1, UGT1A3, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, with UGT2B7 showing the highest activity. Gemfibrozil 54-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 17611564-0 2007 UGT1A1 promoter polymorphism increases risk of nilotinib-induced hyperbilirubinemia. nilotinib 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17611564-3 2007 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) glucuronidates bilirubin in humans, and a polymorphism in the promoter of the gene that encodes it has been associated with hyperbilirubinemia during treatment with a number of drugs. Bilirubin 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-47 17611564-3 2007 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) glucuronidates bilirubin in humans, and a polymorphism in the promoter of the gene that encodes it has been associated with hyperbilirubinemia during treatment with a number of drugs. Bilirubin 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 17927138-5 2007 The results also indicated that well-expressed UGT isoforms in the Caco-2 cells, UGT1A1, UGT1A3, UGT1A6, and UGT2B7, were capable of metabolizing apigenin faster than genistein and that UGT1A6 silencing did not substantially increase the level of expression of genistein-metabolizing UGT isoforms. Genistein 167-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 17927138-5 2007 The results also indicated that well-expressed UGT isoforms in the Caco-2 cells, UGT1A1, UGT1A3, UGT1A6, and UGT2B7, were capable of metabolizing apigenin faster than genistein and that UGT1A6 silencing did not substantially increase the level of expression of genistein-metabolizing UGT isoforms. Genistein 261-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 29793274-7 2007 In that study, a statistically significantly higher level of SN-38 and a relatively lower degree of glucuronidation occurred in patients with the UGT1A1*6 homozygote genotype than in patients with the reference genotype. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 17060921-1 2007 Variants within the human UGT1A1 gene are associated with irinotecan induced severely adverse reactions and hyperbilirubinemia. Irinotecan 58-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 17620344-6 2007 The coexpression of both UGT1A1 and UGT1A4 increased the V(max) values of UGT1A6-catalyzed serotonin and diclofenac O-glucuronide formation. Serotonin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 17620344-6 2007 The coexpression of both UGT1A1 and UGT1A4 increased the V(max) values of UGT1A6-catalyzed serotonin and diclofenac O-glucuronide formation. 1-O-(2-((2',6'-dichlorophenyl)amino)phenylacetyl)glucopyranuronic acid 105-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 17620344-2 2007 The substrates specific for UGT1A1 (estradiol and bilirubin), UGT1A4 (imipramine and trifluoperazine), and UGT1A6 (serotonin and diclofenac) were used to determine the effects of the coexpression of the other UGT1A isoforms on the enzymatic activity. Trifluoperazine 85-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 17620344-3 2007 The coexpression of UGT1A4 and UGT1A6 decreased the S(50) and V(max) values of UGT1A1-catalyzed estradiol 3-O-glucuronide formation and increased the V(max) value of UGT1A1-catalyzed bilirubin O-glucuronide formation. estradiol 3-o-glucuronide 96-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17620344-3 2007 The coexpression of UGT1A4 and UGT1A6 decreased the S(50) and V(max) values of UGT1A1-catalyzed estradiol 3-O-glucuronide formation and increased the V(max) value of UGT1A1-catalyzed bilirubin O-glucuronide formation. Bilirubin 183-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17620344-3 2007 The coexpression of UGT1A4 and UGT1A6 decreased the S(50) and V(max) values of UGT1A1-catalyzed estradiol 3-O-glucuronide formation and increased the V(max) value of UGT1A1-catalyzed bilirubin O-glucuronide formation. o-glucuronide 108-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 17620344-4 2007 The coexpression of UGT1A1 decreased the V(max) value of UGT1A4-catalyzed imipramine N-glucuronide formation but had no effect on UGT1A4-catalyzed trifluoperazine N-glucuronide formation. imipramine N-glucuronide 74-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 17060921-3 2007 Relationship between the concentrations of total serum bilirubin (T-bil) and haplotype structure of UGT1A1 in healthy people were also evaluated. Bilirubin 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 17060921-3 2007 Relationship between the concentrations of total serum bilirubin (T-bil) and haplotype structure of UGT1A1 in healthy people were also evaluated. bil 55-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 17728214-1 2007 The Food and Drug Administration and Pfizer changed the package insert for irinotecan to include a patient"s UGT1A1*28 genotype as a risk factor for severe neutropenia on the basis of the findings of four pharmacogenetic studies, which found that irinotecan-treated patients who were homozygous for the UGT1A1*28 allele had a greater risk of hematologic toxic effects than patients who had one or two copies of the wild-type allele (UGT1A1*1). Irinotecan 75-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17898662-2 2007 Polymorphism of the UGT1A1 gene is one of the likely reasons for interindividual differences in irinotecan pharmacokinetics and severe toxicity. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 121-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 17898662-6 2007 Analysis of genetic variants in genes encoding the drug-metabolizing enzyme (UGT1A1) and transporter (SLCO1B1) involving irinotecan disposition revealed that this patient was homozygous for the SLCO1B1*15 allele, which may result in severe toxicities attributable to the extensive accumulation of SN-38. Irinotecan 297-302 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 17728214-7 2007 The risk of experiencing irinotecan-induced hematologic toxicity for patients with a UGT1A1*28/*28 genotype thus appears to be a function of the dose of irinotecan administered. Irinotecan 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 17728214-1 2007 The Food and Drug Administration and Pfizer changed the package insert for irinotecan to include a patient"s UGT1A1*28 genotype as a risk factor for severe neutropenia on the basis of the findings of four pharmacogenetic studies, which found that irinotecan-treated patients who were homozygous for the UGT1A1*28 allele had a greater risk of hematologic toxic effects than patients who had one or two copies of the wild-type allele (UGT1A1*1). Irinotecan 247-257 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17728214-7 2007 The risk of experiencing irinotecan-induced hematologic toxicity for patients with a UGT1A1*28/*28 genotype thus appears to be a function of the dose of irinotecan administered. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 17697043-0 2007 Substrate-dependent modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1) by propofol in recombinant human UGT1A1 and human liver microsomes. Propofol 78-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-65 17697043-0 2007 Substrate-dependent modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1) by propofol in recombinant human UGT1A1 and human liver microsomes. Propofol 78-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 17697043-0 2007 Substrate-dependent modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1) by propofol in recombinant human UGT1A1 and human liver microsomes. Propofol 78-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 17697043-1 2007 Our previous study has shown that propofol, a probe substrate for human UDP-glucuronosyltransferase (UGT) 1A9, activated the glucuronidation of 4-methylumbelliferone (4-MU) by recombinant UGT1A1 in a concentration-dependent manner. Propofol 34-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 17697043-1 2007 Our previous study has shown that propofol, a probe substrate for human UDP-glucuronosyltransferase (UGT) 1A9, activated the glucuronidation of 4-methylumbelliferone (4-MU) by recombinant UGT1A1 in a concentration-dependent manner. Hymecromone 144-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 17697043-6 2007 In contrast to 4-MUG activity, propofol inhibited UGT1A1-catalysed oestradiol 3beta-glucuronidation in recombinant UGT1A1 as well as in human liver microsomes with IC(50) values of 59 and 228 microM, respectively. Propofol 31-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 17697043-6 2007 In contrast to 4-MUG activity, propofol inhibited UGT1A1-catalysed oestradiol 3beta-glucuronidation in recombinant UGT1A1 as well as in human liver microsomes with IC(50) values of 59 and 228 microM, respectively. Propofol 31-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 17697043-6 2007 In contrast to 4-MUG activity, propofol inhibited UGT1A1-catalysed oestradiol 3beta-glucuronidation in recombinant UGT1A1 as well as in human liver microsomes with IC(50) values of 59 and 228 microM, respectively. Estradiol 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 17697043-6 2007 In contrast to 4-MUG activity, propofol inhibited UGT1A1-catalysed oestradiol 3beta-glucuronidation in recombinant UGT1A1 as well as in human liver microsomes with IC(50) values of 59 and 228 microM, respectively. Estradiol 67-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 17697043-7 2007 In addition, a known UGT1A1 modulator, 17alpha-ethynyloestradiol, stimulated oestradiol 3beta-glucuronidation slightly at a concentration of 5 microM, while it inhibited 4-MUG in recombinant UGT1A1 at all concentrations tested (5-100 microM). Ethinyl Estradiol 39-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 17697043-7 2007 In addition, a known UGT1A1 modulator, 17alpha-ethynyloestradiol, stimulated oestradiol 3beta-glucuronidation slightly at a concentration of 5 microM, while it inhibited 4-MUG in recombinant UGT1A1 at all concentrations tested (5-100 microM). Ethinyl Estradiol 39-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 17697043-7 2007 In addition, a known UGT1A1 modulator, 17alpha-ethynyloestradiol, stimulated oestradiol 3beta-glucuronidation slightly at a concentration of 5 microM, while it inhibited 4-MUG in recombinant UGT1A1 at all concentrations tested (5-100 microM). Estradiol 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 17697043-7 2007 In addition, a known UGT1A1 modulator, 17alpha-ethynyloestradiol, stimulated oestradiol 3beta-glucuronidation slightly at a concentration of 5 microM, while it inhibited 4-MUG in recombinant UGT1A1 at all concentrations tested (5-100 microM). Estradiol 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 191-197 17697043-8 2007 These findings indicate that the modulation of UGT1A1 by propofol is substrate-dependent, and thus care should be taken when extrapolating the stimulatory effects of drugs for one glucuronidation substrate. Propofol 57-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 17627617-7 2007 These exploratory findings suggest that homozygosity for UGT1A1*6 allele may be associated with altered SN-38 disposition and may increase the risk of severe neutropenia in Asian cancer patients, particularly in the Chinese cancer patients who comprised 80% (n = 36) of the patient population in the present study. Irinotecan 104-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 17406868-0 2007 Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6 is associated with reduced activity for SN-38 in Japanese patients with cancer. Irinotecan 103-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 17406868-9 2007 CONCLUSION: Genetic linkage of UGT1A7 and UGT1A9 polymorphisms to UGT1A1*6, related to reduced catalytic and transcriptional activities of UGTs, is associated with the decreased glucuronosyltransferase activity for SN-38 in Japanese patients with cancer. Irinotecan 215-220 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 17591678-9 2007 Collectively, the data indicate that the major mechanism of clearance of raltegravir in humans is UGT1A1-mediated glucuronidation. Raltegravir Potassium 73-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17591679-8 2007 Kinetic and inhibition analyses suggested that the thyroxine glucuronidation in human jejunum microsomes was mainly catalyzed by UGT1A8 and UGT1A10 and to a lesser extent by UGT1A1, and the activity in human kidney microsomes was mainly catalyzed by UGT1A7, UGT1A9, and UGT1A10. Thyroxine 51-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 17576806-0 2007 Stereoselective glucuronidation of 5-(4"-hydroxyphenyl)-5-phenylhydantoin by human UDP-glucuronosyltransferase (UGT) 1A1, UGT1A9, and UGT2B15: effects of UGT-UGT interactions. hydroxyphenytoin 35-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-120 17576806-6 2007 Among human UDP-glucuronosyltransferase (UGT) enzymes, UGT1A1, UGT1A9, and UGT2B15 showed 4"-HPPH O-glucuronide formation. o-glucuronide 98-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 17478602-9 2007 The correlation of UGT1A1 with total bilirubin levels was mainly due to (TA)(n) repeats in Caucasians but a clear correlation was not observed in African Americans because of the high diversity of haplotypes and the small sample size. Bilirubin 37-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 17576806-7 2007 Interestingly, UGT1A1 stereoselectively formed (R)-4"-HPPH O-glucuronide, whereas UGT1A9 and UGT2B15 stereoselectively formed (S)-4"-HPPH O-glucuronide from racemic 4"-HPPH. (r)-4"-hpph o-glucuronide 47-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17576806-7 2007 Interestingly, UGT1A1 stereoselectively formed (R)-4"-HPPH O-glucuronide, whereas UGT1A9 and UGT2B15 stereoselectively formed (S)-4"-HPPH O-glucuronide from racemic 4"-HPPH. hydroxyphenytoin 51-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17576806-9 2007 It was demonstrated that coexpression of UGT1A4 increased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A1 but decreased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A9. Sulfur 79-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 17576806-9 2007 It was demonstrated that coexpression of UGT1A4 increased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A1 but decreased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A9. (r)-4"-hpph o-glucuronide 88-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 17576806-9 2007 It was demonstrated that coexpression of UGT1A4 increased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A1 but decreased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A9. Sulfur 179-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 17576806-9 2007 It was demonstrated that coexpression of UGT1A4 increased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A1 but decreased the V(max) values of (S)- and (R)-4"-HPPH O-glucuronide formation catalyzed by UGT1A9. (r)-4"-hpph o-glucuronide 188-213 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 17576806-10 2007 Coexpression of UGT1A6 increased the S(50) values and decreased the V(max) values of (S)- and (R)-4"-HPPH glucuronide formation catalyzed by UGT1A1 and UGT1A9. 3-azido-2,7-naphthalene disulfonate 85-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 17576806-10 2007 Coexpression of UGT1A6 increased the S(50) values and decreased the V(max) values of (S)- and (R)-4"-HPPH glucuronide formation catalyzed by UGT1A1 and UGT1A9. (r)-4"-hpph glucuronide 94-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 17576806-12 2007 In conclusion, we found that 4"-HPPH O-glucuronide formation in human liver microsomes is catalyzed by UGT1A1, UGT1A9, and UGT2B15 in a stereoselective manner, being modulated by interaction with other UGT1A isoforms. 4"-hpph o-glucuronide 29-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 17691917-5 2007 So far, the candidate gene approach has provided important clues for pharmacogenomic-based personalized chemotherapy with 6-mercaptopurine (6-MP), solely metabolized by thiopurine S-methyltransferase (TPMT), and irinotecan, mainly detoxified by UDP-glucuronosyltransferase 1A1 (UGT1A1). Mercaptopurine 122-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 245-276 17691917-5 2007 So far, the candidate gene approach has provided important clues for pharmacogenomic-based personalized chemotherapy with 6-mercaptopurine (6-MP), solely metabolized by thiopurine S-methyltransferase (TPMT), and irinotecan, mainly detoxified by UDP-glucuronosyltransferase 1A1 (UGT1A1). Mercaptopurine 122-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 278-284 17691917-5 2007 So far, the candidate gene approach has provided important clues for pharmacogenomic-based personalized chemotherapy with 6-mercaptopurine (6-MP), solely metabolized by thiopurine S-methyltransferase (TPMT), and irinotecan, mainly detoxified by UDP-glucuronosyltransferase 1A1 (UGT1A1). Mercaptopurine 140-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 245-276 17691917-5 2007 So far, the candidate gene approach has provided important clues for pharmacogenomic-based personalized chemotherapy with 6-mercaptopurine (6-MP), solely metabolized by thiopurine S-methyltransferase (TPMT), and irinotecan, mainly detoxified by UDP-glucuronosyltransferase 1A1 (UGT1A1). Mercaptopurine 140-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 278-284 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Mercaptopurine 158-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Mercaptopurine 158-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 17691917-6 2007 Reduced activity of TPMT caused by polymorphisms in the TPMT gene and decreased activity of UGT1A1 caused by UGT1A1*28 are related to severe toxic effects of 6-MP and irinotecan, respectively. Irinotecan 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 17628876-4 2007 The phenolic glucuronidation of the curcuminoids was predominantly catalyzed by hepatic UGT1A1 and intestinal UGT1A8 and 1A10, whereas UGT1A9, 2B7, and 1A8 exhibited high activities for hexahydro-curcuminoids. curcuminoids 36-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 17622938-0 2007 UGT1A1*28 genotype affects the in-vitro glucuronidation of thyroxine in human livers. Thyroxine 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17622938-6 2007 RESULTS: A strong correlation was observed between the glucuronidation of T4 and SN-38, a UGT1A1 substrate (r=0.82, P<0.0001). Irinotecan 81-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 17622938-7 2007 A significant trend of decreasing T4 glucuronide (T4G) levels was observed with increasing number of UGT1A1 -53(TA)7 alleles (P=0.001). t4 glucuronide 34-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 17593033-5 2007 Regression analysis showed that serum bilirubin levels and the incidence of gallstones were strongly associated with the number of UGT1A1 [TA] repeats in all subjects (P < 0.0001 and P < 0.01, respectively). Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 17498780-7 2007 These results showed that AhR, UGT1A1, GSTP1 and GSTT1 polymorphisms were associated with urinary 1-OHP concentrations in Chinese coke oven workers. Oxaliplatin 98-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 17762398-2 2007 The presence of an additional TA repeat in the TATA sequence of the UGT1A1 promoter leads to a significant decrease in SN-38 glucuronidation. Irinotecan 119-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 17762398-3 2007 Patients with the UGT1A1 (TA)7 allele are more likely to experience severe neutropenia and diarrhea following irinotecan chemotherapy. Irinotecan 110-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 17593033-1 2007 Serum bilirubin levels and predisposition to gallstones in sickle cell disease (SCD) are influenced by genetic variation in the hepatic uridine diphosphate (UDP)-glucuronosyltransferase (UGT1A1) gene, but the association is not consistent. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17558305-10 2007 CONCLUSIONS: The haplotypes significantly associated with reduced area under concentration curve ratios and neutropenia contained UGT1A1*6 or *28, and both of them should be genotyped before irinotecan is given to Japanese and probably other Asian patients. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 17593033-1 2007 Serum bilirubin levels and predisposition to gallstones in sickle cell disease (SCD) are influenced by genetic variation in the hepatic uridine diphosphate (UDP)-glucuronosyltransferase (UGT1A1) gene, but the association is not consistent. hepatic uridine diphosphate 128-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17593033-1 2007 Serum bilirubin levels and predisposition to gallstones in sickle cell disease (SCD) are influenced by genetic variation in the hepatic uridine diphosphate (UDP)-glucuronosyltransferase (UGT1A1) gene, but the association is not consistent. Uridine Diphosphate 157-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17681105-1 2007 BACKGROUND: Uridine diphosphate glucuronosyltransferase (UGT) 1A1 7/7 polymorphism has been linked with an increased risk of irinotecan-induced severe toxicities. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-65 17681105-2 2007 We evaluated UGT1A1 polymorphism in patients developing grade 3/4 toxicity after initiation of irinotecan to determine the frequency of this polymorphism in this population. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 17681105-3 2007 PATIENTS AND METHODS: Twenty patients with grade 3/4 irinotecan-induced toxicity underwent UGT1A1 genotyping in an exploratory study. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-97 17681105-8 2007 Our data support the need for more prospective studies that evaluate the predictive value of UGT1A1 as well as UGT1A1-based dosing in patients receiving irinotecan. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 17681105-8 2007 Our data support the need for more prospective studies that evaluate the predictive value of UGT1A1 as well as UGT1A1-based dosing in patients receiving irinotecan. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 17599282-3 2007 In particular, UGT1A1-dependent bilirubin conjugation plays a critical role in the detoxification of neurotoxic bilirubin and phenobarbital-mediated UGT1A1 induction therapy is commonly used in the treatment of unconjugated hyperbilirubinemic diseases such as Crigler-Najjar type II disease. Bilirubin 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17599282-3 2007 In particular, UGT1A1-dependent bilirubin conjugation plays a critical role in the detoxification of neurotoxic bilirubin and phenobarbital-mediated UGT1A1 induction therapy is commonly used in the treatment of unconjugated hyperbilirubinemic diseases such as Crigler-Najjar type II disease. Bilirubin 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 17599282-3 2007 In particular, UGT1A1-dependent bilirubin conjugation plays a critical role in the detoxification of neurotoxic bilirubin and phenobarbital-mediated UGT1A1 induction therapy is commonly used in the treatment of unconjugated hyperbilirubinemic diseases such as Crigler-Najjar type II disease. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17599282-3 2007 In particular, UGT1A1-dependent bilirubin conjugation plays a critical role in the detoxification of neurotoxic bilirubin and phenobarbital-mediated UGT1A1 induction therapy is commonly used in the treatment of unconjugated hyperbilirubinemic diseases such as Crigler-Najjar type II disease. Phenobarbital 126-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17599282-3 2007 In particular, UGT1A1-dependent bilirubin conjugation plays a critical role in the detoxification of neurotoxic bilirubin and phenobarbital-mediated UGT1A1 induction therapy is commonly used in the treatment of unconjugated hyperbilirubinemic diseases such as Crigler-Najjar type II disease. Phenobarbital 126-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 17446261-4 2007 UGT1A1, 1A3, 1A9, 2B4, and 2B7 showed glucuronidation activity for both (R)- and (S)-glucuronide, with UGT2B7 possessing the highest activity. (r)- and (s)-glucuronide 72-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Pyranocoumarins 63-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Pyranocoumarins 63-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 351-357 17426648-1 2007 Increased heme catabolism has been reported in glucose-6-phosphate dehydrogenase (G-6-PD)-normal neonates who were also homozygous for (TA)7/(TA)7 (UGT1A1*28) uridine diphosphoglucuronate-glucuronosyltransferase 1A1 (UGT) promoter polymorphism (Gilbert syndrome). Heme 10-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. CHEMBL2334004 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. cis-avicennol 93-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. cis-avicennol 93-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 351-357 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Furocoumarins 116-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Furocoumarins 116-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 351-357 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Coumarins 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. Coumarins 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 351-357 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. coumarin 69-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 17599282-5 2007 After 3 days of treatment at a concentration of 25 microM, the pyranocoumarins avicennin and CIS-avicennol, and the furocoumarins bergapten and imperatorin, increased by 2-fold UGT1A1-dependent activity, equivalent to the increases obtained with chrysin at 25 microM, whereas in the presence of the simple coumarins such as coumarin or umbelliferone, UGT1A1-dependent activity was not modified. coumarin 69-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 351-357 17599282-6 2007 In terms of structural requirements for UGT1A1 induction, the present study suggests that the B-ring (phenyl) for chrysin and the furan or pyran rings for coumarins are essential for the biological activity. furan 130-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 17599282-6 2007 In terms of structural requirements for UGT1A1 induction, the present study suggests that the B-ring (phenyl) for chrysin and the furan or pyran rings for coumarins are essential for the biological activity. Pyrans 139-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 17599282-6 2007 In terms of structural requirements for UGT1A1 induction, the present study suggests that the B-ring (phenyl) for chrysin and the furan or pyran rings for coumarins are essential for the biological activity. Coumarins 155-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 17577039-0 2007 UGT1A1 promoter genotype correlates with SN-38 pharmacokinetics, but not severe toxicity in patients receiving low-dose irinotecan. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17608024-0 2007 UGT1AI*6 and UGT1A1*27 for individualized irinotecan chemotherapy. Irinotecan 42-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 13-19 17608024-1 2007 Genetic polymorphisms of uridine 5"-diphospho-glucuronosyl-transferase (UGT)1A1, a crucial drug-metabolizing enzyme of the anticancer drug irinotecan, are essential determinants of individual variation in susceptibility to irinotecan-related toxicity. Uridine 25-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-79 17608024-1 2007 Genetic polymorphisms of uridine 5"-diphospho-glucuronosyl-transferase (UGT)1A1, a crucial drug-metabolizing enzyme of the anticancer drug irinotecan, are essential determinants of individual variation in susceptibility to irinotecan-related toxicity. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-79 17608024-1 2007 Genetic polymorphisms of uridine 5"-diphospho-glucuronosyl-transferase (UGT)1A1, a crucial drug-metabolizing enzyme of the anticancer drug irinotecan, are essential determinants of individual variation in susceptibility to irinotecan-related toxicity. Irinotecan 223-233 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-79 17608024-4 2007 Therefore, it is reasonable to assume that the presence of UGT1A1*6 or UGTIA1*27 would also increase the risk of irinotecan toxicity. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 17510208-0 2007 UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Irinotecan 78-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17475008-8 2007 Molecular docking experiments identified key residues in donor and acceptor recognition and provided insight into the catalytic mechanism of UGT glucuronidation, suggesting the human UGT1A1 residue histidine 39 (H39) as a general base and the residue aspartic acid 151 (D151) as an important electron-transfer helper. Histidine 198-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17529881-4 2007 SN-38 is further metabolized to SN-38G by various hepatic and extrahepatic UGT1A isozymes, mainly UGT1A1. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 17529881-5 2007 Impaired glucuronidation activity of the UGT1A1 enzyme has been linked with elevated levels of SN-38, leading to toxicities. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 17529881-6 2007 UGT1A1*28 involves an extra TA repeat in the UGT1A1 promoter region and is the variant most frequently contributing to interpatient variability in irinotecan pharmacokinetics and toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17529881-6 2007 UGT1A1*28 involves an extra TA repeat in the UGT1A1 promoter region and is the variant most frequently contributing to interpatient variability in irinotecan pharmacokinetics and toxicities. Irinotecan 147-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 17529881-8 2007 Recently, UGT1A1*6 seems to contribute to the risk of toxicity of irinotecan in Asian patients. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 17293379-6 2007 Using the single expression systems, it was confirmed that estradiol 3-O-glucuronide, imipramine N-glucuronide, serotonin O-glucuronide, and propofol O-glucuronide formations are specific for UGT1A1, UGT1A4, UGT1A6, and UGT1A9, respectively. estradiol 3-o-glucuronide 59-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 17293379-6 2007 Using the single expression systems, it was confirmed that estradiol 3-O-glucuronide, imipramine N-glucuronide, serotonin O-glucuronide, and propofol O-glucuronide formations are specific for UGT1A1, UGT1A4, UGT1A6, and UGT1A9, respectively. imipramine N-glucuronide 86-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 17293379-6 2007 Using the single expression systems, it was confirmed that estradiol 3-O-glucuronide, imipramine N-glucuronide, serotonin O-glucuronide, and propofol O-glucuronide formations are specific for UGT1A1, UGT1A4, UGT1A6, and UGT1A9, respectively. serotonin o-glucuronide 112-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 17293379-6 2007 Using the single expression systems, it was confirmed that estradiol 3-O-glucuronide, imipramine N-glucuronide, serotonin O-glucuronide, and propofol O-glucuronide formations are specific for UGT1A1, UGT1A4, UGT1A6, and UGT1A9, respectively. propofol o-glucuronide 141-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 17293379-8 2007 On the other hand, the coexpressed UGT1A1 increased Km and decreased the Vmax of the propofol O-glucuronide formation catalyzed by UGT1A9. Propofol 85-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 17293379-8 2007 On the other hand, the coexpressed UGT1A1 increased Km and decreased the Vmax of the propofol O-glucuronide formation catalyzed by UGT1A9. o-glucuronide 94-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 17347060-1 2007 BACKGROUND AND AIM: To clarify the precise mode of inheritance of Gilbert syndrome, an unconjugated familial hyperbilirubinemia, where impaired bilirubin conjugation is caused by reduced UGT1A1 activity determined by a defective function of the A(TA)6TAA promoter region of the UGT1A1 gene. Bilirubin 114-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17220234-9 2007 Tranilast glucuronosyltransferase activity was strongly inhibited by bilirubin, a typical UGT1A1 substrate, and N-3, indicating that the phase I metabolite could affect the tranilast glucuronosyltransferase activity. Bilirubin 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 17347060-1 2007 BACKGROUND AND AIM: To clarify the precise mode of inheritance of Gilbert syndrome, an unconjugated familial hyperbilirubinemia, where impaired bilirubin conjugation is caused by reduced UGT1A1 activity determined by a defective function of the A(TA)6TAA promoter region of the UGT1A1 gene. Bilirubin 114-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 278-284 17374650-0 2007 UGT1A1 polymorphism is associated with serum bilirubin concentrations in a randomized, controlled, fruit and vegetable feeding trial. Bilirubin 45-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 17374650-1 2007 UDP-glucuronosyltransferase (UGT) 1A1 glucuronidates bilirubin, estrogens, and exogenous compounds, including dietary carcinogens. Bilirubin 53-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 17374650-2 2007 The UGT1A1*28 polymorphism, characterized by variation in the number of thymine-adenine repeats in the promoter region, modulates UGT1A1 transcription. thymine-adenine 72-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 17374650-2 2007 The UGT1A1*28 polymorphism, characterized by variation in the number of thymine-adenine repeats in the promoter region, modulates UGT1A1 transcription. thymine-adenine 72-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 130-136 17496722-2 2007 The most common genotype of Gilbert"s syndrome is the homozygous polymorphism, A(TA)7TAA, in the promoter of the gene for UDP-glucuronosyltransferase 1A1 (UGT1A1), with a thymine adenine insertion in the TATA-box-like sequence, which results in a decrease in UGT1A1 activity. thymine adenine 171-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-153 17496722-2 2007 The most common genotype of Gilbert"s syndrome is the homozygous polymorphism, A(TA)7TAA, in the promoter of the gene for UDP-glucuronosyltransferase 1A1 (UGT1A1), with a thymine adenine insertion in the TATA-box-like sequence, which results in a decrease in UGT1A1 activity. thymine adenine 171-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 17496722-2 2007 The most common genotype of Gilbert"s syndrome is the homozygous polymorphism, A(TA)7TAA, in the promoter of the gene for UDP-glucuronosyltransferase 1A1 (UGT1A1), with a thymine adenine insertion in the TATA-box-like sequence, which results in a decrease in UGT1A1 activity. thymine adenine 171-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 259-265 17151191-0 2007 UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Etoposide 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 17196409-2 2007 The causative mutation in Caucasians is almost exclusively a TA dinucleotide insertion in the TATA box of the UGT1A1 promoter. Dinucleoside Phosphates 64-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 17471158-3 2007 This results in a reduced UGT 1A1 expression with 70% less glucuronidation capacity for bilirubin and other UGT1A1 substrates. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-33 17471158-6 2007 The anticancer agent irinotecan is metabolized to the active SN-38, which is further glucuronidated and detoxified by UGT 1A1. Irinotecan 21-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-125 17471158-6 2007 The anticancer agent irinotecan is metabolized to the active SN-38, which is further glucuronidated and detoxified by UGT 1A1. Irinotecan 61-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-125 17259171-3 2007 When human HepG2 cells were treated with the prooxidants tert-butylhydroquinone and beta-naphthoflavone, cellular UGT1A1 glucuronidation activities were increased. 2-tert-butylhydroquinone 57-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 17259171-3 2007 When human HepG2 cells were treated with the prooxidants tert-butylhydroquinone and beta-naphthoflavone, cellular UGT1A1 glucuronidation activities were increased. beta-Naphthoflavone 84-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 17259171-8 2007 Gene mapping experiments including transfections of UGT1A1 reporter gene constructs into HepG2 cells coupled with functional analysis of Nrf2 expression and binding to anti-oxidant-response elements (ARE) resulted in identification of an ARE in the phenobarbital-response enhancer module region of the UGT1A1 gene. Phenobarbital 249-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 17259171-8 2007 Gene mapping experiments including transfections of UGT1A1 reporter gene constructs into HepG2 cells coupled with functional analysis of Nrf2 expression and binding to anti-oxidant-response elements (ARE) resulted in identification of an ARE in the phenobarbital-response enhancer module region of the UGT1A1 gene. Phenobarbital 249-262 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 302-308 16897040-8 2007 Phenolphthalein, a substrate for UGT1A9, inhibited DL glucuronidation in HLM competitively (K (i) = 0.356 mM), but bilirubin, a substrate for UGT1A1, did not. Phenolphthalein 0-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 17531109-0 2007 Reversible grade 4 hyperbilirubinemia in a patient with UGT1A1 7/7 genotype treated with irinotecan and cetuximab. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 17531109-5 2007 This case outlines the possibility of severe hepatic toxicity with moderate doses of irinotecan in patients with a UGT1A1 7/7 genotype. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 17151191-0 2007 UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Glucuronides 194-206 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 17151191-0 2007 UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Etoposide 210-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 17151191-3 2007 In vitro UDP-glucuronosyltransferase (UGT) reaction screening with 12 recombinant human UGTs demonstrated that etoposide glucuronidation is mainly catalyzed by UGT1A1. Etoposide 111-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 160-166 17151191-4 2007 Although UGT1A8 and 1A3 also catalyzed the glucuronidation of etoposide, their activities were approximately 10 and 1% of UGT1A1. Etoposide 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 17151191-9 2007 The data indicated that UGT1A1 is principally responsible for the formation of etoposide glucuronides, mainly in the form of phenolic glucuronide, suggesting that etoposide can be used as a highly selective probe substrate for human UGT1A1 in vitro. etoposide glucuronide 79-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 17245571-4 2007 RESULTS: Among the UGT isozymes investigated, UGT1A1, 1A3, 1A9, and 2B7 showed glucuronidation activity for indomethacin, with UGT1A9 possessing the highest activity, followed by UGT2B7. Indomethacin 108-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 17151191-9 2007 The data indicated that UGT1A1 is principally responsible for the formation of etoposide glucuronides, mainly in the form of phenolic glucuronide, suggesting that etoposide can be used as a highly selective probe substrate for human UGT1A1 in vitro. etoposide glucuronide 79-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 233-239 17151191-9 2007 The data indicated that UGT1A1 is principally responsible for the formation of etoposide glucuronides, mainly in the form of phenolic glucuronide, suggesting that etoposide can be used as a highly selective probe substrate for human UGT1A1 in vitro. Glucuronides 89-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 17151191-9 2007 The data indicated that UGT1A1 is principally responsible for the formation of etoposide glucuronides, mainly in the form of phenolic glucuronide, suggesting that etoposide can be used as a highly selective probe substrate for human UGT1A1 in vitro. Etoposide 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 17151191-9 2007 The data indicated that UGT1A1 is principally responsible for the formation of etoposide glucuronides, mainly in the form of phenolic glucuronide, suggesting that etoposide can be used as a highly selective probe substrate for human UGT1A1 in vitro. Etoposide 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 233-239 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Fluorouracil 323-337 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Fluorouracil 339-343 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Irinotecan 350-360 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Oxaliplatin 364-375 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17273745-1 2007 The aim of this study was to investigate the influence of combining thymidylate synthase (TS), X-ray cross complementing factor 1 (XRCC1) and uridine diphosphate glucoronosyltransferase (UGT1A1 *28) polymorphism genotypes in response rate and time to progression (TTP) in metastatic colorectal cancer patients treated with 5-fluorouracil (5-FU) plus irinotecan or oxaliplatin (OXA). Oxaliplatin 377-380 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 17259247-0 2007 A combinatorial haplotype of the UDP-glucuronosyltransferase 1A1 gene (#60-#IB) increases total bilirubin concentrations in Japanese volunteers. Bilirubin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-64 17166930-7 2007 We investigated the frequency of promoter region [thymine-adenine(TA)]7 polymorphism in UGT1A1 gene. thymine-adenine 50-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 17108060-14 2007 The induction effect of catechins on UGT1A1 seems to be modest and highly variable. Catechin 24-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 17256720-1 2007 UNLABELLED: The flavonoid chrysin is an important dietary substance and induces UGT1A1 protein expression in cell culture. Flavonoids 16-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 17256720-2 2007 As a representative of the class of dietary flavonoids, clinical investigations have been considered as a means of inducing hepatic UGT1A1 expression. Flavonoids 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 17256720-5 2007 However, key differences in Ah receptor recognition and activation of UGT1A1 by chrysin exist when compared with classical mechanisms of UGT1A1 induction by TCDD. Polychlorinated Dibenzodioxins 157-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 17410034-3 2007 Genetic polymorphisms in the UGT1A1 gene have been identified as one of the causes of the increased diarrhea seen in white patients treated with irinotecan when compared with Japanese patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 17166930-7 2007 We investigated the frequency of promoter region [thymine-adenine(TA)]7 polymorphism in UGT1A1 gene. Tantalum 66-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 17148966-2 2007 Polymorphisms at the uridin-glucoronosyl-transferase 1A1 (UGT1A1) and multidrug resistance 1 (MDR1) genes may influence, respectively, bilirubin and ATV plasma concentrations. Bilirubin 135-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-56 17484518-6 2007 Docking of UDP-glucuronic acid to a model of UGT1A1 resulted in a root mean square deviation of only 0.37 angstroms vs. UDP co-crystallized with the plant UGT71G1 template. Uridine Diphosphate Glucuronic Acid 11-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 17484518-6 2007 Docking of UDP-glucuronic acid to a model of UGT1A1 resulted in a root mean square deviation of only 0.37 angstroms vs. UDP co-crystallized with the plant UGT71G1 template. Uridine Diphosphate 11-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 17484518-7 2007 The significance of a comparative model generated for UGT1A1 with respect to both the sugar donor and aglycone binding sites, and mechanism, is discussed. Sugars 86-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 17484518-7 2007 The significance of a comparative model generated for UGT1A1 with respect to both the sugar donor and aglycone binding sites, and mechanism, is discussed. CHEBI:166892 102-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 17148966-2 2007 Polymorphisms at the uridin-glucoronosyl-transferase 1A1 (UGT1A1) and multidrug resistance 1 (MDR1) genes may influence, respectively, bilirubin and ATV plasma concentrations. Bilirubin 135-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 17148966-2 2007 Polymorphisms at the uridin-glucoronosyl-transferase 1A1 (UGT1A1) and multidrug resistance 1 (MDR1) genes may influence, respectively, bilirubin and ATV plasma concentrations. Atazanavir Sulfate 149-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-56 17148966-2 2007 Polymorphisms at the uridin-glucoronosyl-transferase 1A1 (UGT1A1) and multidrug resistance 1 (MDR1) genes may influence, respectively, bilirubin and ATV plasma concentrations. Atazanavir Sulfate 149-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 17187418-0 2007 Regulation of the UGT1A1 bilirubin-conjugating pathway: role of a new splicing event at the UGT1A locus. Bilirubin 25-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 17185998-2 2007 Life-threatening diarrhea is observed in up to 25% of patients receiving irinotecan and has been related with irinotecan pharmacokinetics and UGT1A1 genotype status. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 17185998-10 2007 As the association was seen in patients not genetically predisposed at risk for diarrhea due to UGT1A1*28, confirmatory studies of the relationships of ABCC2 genotypes and irinotecan disposition and toxicity are warranted. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 19356014-0 2007 ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Irinotecan 139-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 17185519-4 2006 The irinotecan/UGT1A1 issue and the development of molecular diagnostic testing are now to be translated into clinical practice. Irinotecan 4-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 17264799-1 2007 BACKGROUND: Glucuronidation by the UDP glucuronosyltransferase 1A enzymes (UGT1As) is a major pathway for elimination of drugs and endogenous substances, such as bilirubin. Bilirubin 162-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-65 17184208-1 2006 Depending upon the UDP glucuronosyltransferase 1A1 (UGT1A1) genotype, patients are more or less susceptible to the risk of severe toxicity of irinotecan. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-50 17184208-1 2006 Depending upon the UDP glucuronosyltransferase 1A1 (UGT1A1) genotype, patients are more or less susceptible to the risk of severe toxicity of irinotecan. Irinotecan 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 17184208-2 2006 As the US FDA-approved label of irinotecan (CPT-11, Camptosar) has been recently revised to include UGT1A1 genotype among potential risk factors for toxicity, it is expected that UGT1A1 genotyping will be increasingly used in patients undergoing irinotecan treatment. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 17090741-5 2006 The labeling of irinotecan was recently changed and now includes a warning of greater neutropenia risk in patients with reduced activity in the drug-metabolizing enzyme uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-216 17090741-5 2006 The labeling of irinotecan was recently changed and now includes a warning of greater neutropenia risk in patients with reduced activity in the drug-metabolizing enzyme uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 218-224 17090741-7 2006 Numerous studies have demonstrated the effects of genetic factors, especially UGT1A1*28, that contribute to interpatient variability in irinotecan pharmacokinetics and toxicity. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 17090741-8 2006 Irinotecan"s new labeling recommends that clinicians consider reducing the dosage of irinotecan in patients homozygous for UGT1A1*28. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 17090741-8 2006 Irinotecan"s new labeling recommends that clinicians consider reducing the dosage of irinotecan in patients homozygous for UGT1A1*28. Irinotecan 85-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 17090741-9 2006 CONCLUSION: At least part of the interpatient variability of irinotecan toxicity can be explained by the UGT1A1*28 polymorphism. Irinotecan 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 17090741-10 2006 Patients who are homozygous for the UGT1A1*28 allele have an increased risk of developing severe neutropenia when receiving irinotecan, especially the 300-350- mg/m2 regimen. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 17185519-1 2006 This article focuses on pharmacogenetic associations between genetic polymorphism of uridine diphosphate glucuronosyltransferase (UGT) 1A1 gene and irinotecan toxicity. Irinotecan 148-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-138 17185519-3 2006 On the basis of these backgrounds, the irinotecan label was updated in 2005 in the United States to provide pharmacogenetic information, and a dose reduction of irinotecan should be considered for patients known to be homozygous for the UGT1A1*28 allele when administered in combination with other agents or a single agent. Irinotecan 161-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 237-243 17192505-3 2007 This analysis extended the work of a previous study that examined the effect of UGT1A1 genotypes on the incidence of severe neutropenia in 86 advanced cancer patients following irinotecan treatment. Irinotecan 177-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 17192505-7 2007 Based on these findings and other reports, the irinotecan label was modified to indicate the role of UGT1A1*28 polymorphism in the metabolism of irinotecan and the associated increased risk of severe neutropenia. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 17192505-7 2007 Based on these findings and other reports, the irinotecan label was modified to indicate the role of UGT1A1*28 polymorphism in the metabolism of irinotecan and the associated increased risk of severe neutropenia. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 17192505-8 2007 The label modifications also included recommendations for lower starting doses of irinotecan in patients homozygous for the UGT1A1*28 (7/7) polymorphism. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 17164591-6 2007 Patients who have UGT1A6*2 without UGT1A1*28 showed a significant, lower area under concentration-time curve (AUC(0)-->infinity) of PCM, PCM-glucuronide and PCM-sulfate than those of the patients with wild-type UGT1A1 and UGT1A6 (p < 0.05). acetaminophen glucuronide 140-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 17164591-6 2007 Patients who have UGT1A6*2 without UGT1A1*28 showed a significant, lower area under concentration-time curve (AUC(0)-->infinity) of PCM, PCM-glucuronide and PCM-sulfate than those of the patients with wild-type UGT1A1 and UGT1A6 (p < 0.05). acetaminophen sulfate ester 160-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 17090120-5 2006 Supersomes containing human UGT1A1 and 1A3 exclusively generated the phenolic glucuronide, albeit with very low activities, whereas UGT1A9 catalyzed the specific formation of the alcoholic glucuronide and UGT2B7 the predominant formation of the phenolic glucuronide with high activities. Glucuronides 78-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-42 17090120-5 2006 Supersomes containing human UGT1A1 and 1A3 exclusively generated the phenolic glucuronide, albeit with very low activities, whereas UGT1A9 catalyzed the specific formation of the alcoholic glucuronide and UGT2B7 the predominant formation of the phenolic glucuronide with high activities. Glucuronides 189-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-42 17090120-5 2006 Supersomes containing human UGT1A1 and 1A3 exclusively generated the phenolic glucuronide, albeit with very low activities, whereas UGT1A9 catalyzed the specific formation of the alcoholic glucuronide and UGT2B7 the predominant formation of the phenolic glucuronide with high activities. Glucuronides 189-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-42 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-0 2006 Pharmacogenetic impact of polymorphisms in the coding region of the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with cancer. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 204-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-1 2006 Pharmacogenetic testing for UDP-glucuronosyltransferase (UGT) 1A1*28, a promoter variant of the UGT1A1 gene, is now carried out clinically to estimate the risk of irinotecan-associated toxicity. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-1 2006 Pharmacogenetic testing for UDP-glucuronosyltransferase (UGT) 1A1*28, a promoter variant of the UGT1A1 gene, is now carried out clinically to estimate the risk of irinotecan-associated toxicity. Irinotecan 163-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 174-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). Irinotecan 186-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 146-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 16965601-4 2006 Pharmacogenetic relationships were explored between the UGT1A1 genotype and the ratio of the area under the plasma concentration-time curve (AUC) of the active metabolite of irinotecan (SN-38) to that of SN-38 glucuronide (SN-38G), used as a surrogate for UGT1A1 activity (AUC(SN-38)/AUC(SN-38G)). 7-ethyl-10-hydroxycamptothecin glucuronide 204-221 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16965601-10 2006 The two patients simultaneously heterozygous for UGT1A1*28 and UGT1A1*6 and the two patients homozygous for UGT1A1*6 had significantly higher AUC(SN-38)/AUC(SN-38G) ratios than the others (P = 0.0039). Irinotecan 157-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-11 2006 Concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, altered the disposition of irinotecan remarkably, potentially increasing susceptibility to toxicity. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 16965601-11 2006 Concurrence of UGT1A1*28 and UGT1A1*6, even when heterozygous, altered the disposition of irinotecan remarkably, potentially increasing susceptibility to toxicity. Irinotecan 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16965601-13 2006 UGT1A1 polymorphisms in the coding region of the UGT1A1 gene should be genotyped in addition to testing for UGT1A1*28 to more accurately predict irinotecan-related toxicity, at least in Asian patients. Irinotecan 145-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 17315542-13 2006 The formation of DRF-6574 glucuronide by human liver, intestinal and UGT1A1, 1A3 and 1A8 microsomes was effectively inhibited by phenylbutazone. Glucuronides 26-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 17058217-1 2006 Gilbert"s disease leads to intermittent non-hemolytic hyperbilirubinemia by a reduction of hepatic bilirubin glucuronidation associated with the presence of the UDP-glucuronosyltransferase (UGT) 1A1*28 polymorphism. Bilirubin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-198 17058217-9 2006 Homozygous combinations of UGT1A1*28 with the other variants increased from 7.4% (normal bilirubin to 42 micromol/L) to 41% to 46.1% (43 to >85 micromol/L), and 100% (>85 micromol/L). Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 29788595-0 2006 Novel personalized medicine technology: UGT1A1 testing for irinotecan as a case study. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-53 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 29788595-1 2006 Third Wave"s Invader UDP glucuronosyltransferase 1A1 (UGT1A1) Molecular Assay, a genotyping system to predict adverse drug reactions in patients receiving the chemotherapeutic agent irinotecan (Camptosar , Pzifer, NY, USA) for the treatment of metastatic colorectal cancer (mCRC), was recently approved by the US FDA. Irinotecan 195-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 29788595-3 2006 This descriptive analysis highlight these data and the issues for the translation of this test to practice, including gaps in the evidence base, issues regarding adoption of the test to clinical practice and the potential societal impact of UGT1A1 testing for irinotecan prescribing. Irinotecan 260-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 241-247 16982469-3 2006 Recent FDA approval of genetic testing for mutations in the UGT1A1 gene that predict adverse reactions to irinotecan is ushering in a new era that will increasingly rely on genotyping to individualize treatment decisions for patients with cancer as well as for patients at high risk who may be candidates for chemoprevention agents. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. doxifluridine 151-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. doxifluridine 151-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Fluorouracil 396-400 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Fluorouracil 396-400 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 17016579-3 2006 We present a colon cancer patient with the UGT1A1 polymorphism (UGT1A1 *28) as a known high risk for irinotecan, who was treated with a combination of doxifluridine and irinotecan for peritoneal dissemination resulting in stable disease for 2 years without adverse reactions, although the patient initially developed severe adverse effects to the combination of the protracted venous infusion of 5-FU and irinotecan. Irinotecan 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 17009167-13 2006 Furthermore, we show that after deglycosylation, the resulting aglycone is metabolized in trans-resveratrol-3-O-ss-glucuronide and to a lesser extent in trans-resveratrol-4"-O-ss-glucuronide, and that UGT1A1 is mainly involved in this metabolism. CHEBI:166892 63-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 201-207 17315542-13 2006 The formation of DRF-6574 glucuronide by human liver, intestinal and UGT1A1, 1A3 and 1A8 microsomes was effectively inhibited by phenylbutazone. Phenylbutazone 129-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 16791115-1 2006 OBJECTIVE: The aim of the study was to investigate relationships among indinavir, lamivudine-triphosphate, and zidovudine-triphosphate pharmacokinetics and pharmacodynamics with polymorphisms in CYP3A5, MDR1, MRP2, MRP4, BCRP, and UGT1A1 genes. zidovudine triphosphate 111-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 16545899-4 2006 7-Ethyl-10-hydroxycamptothecin (SN-38) glucuronidation, used as a probe for UGT1A1, showed sigmoidal kinetics with a Hill coefficient (n) of 1.2-1.3 in control and BNF-pretreated HepG2 cells. Irinotecan 0-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 16545899-4 2006 7-Ethyl-10-hydroxycamptothecin (SN-38) glucuronidation, used as a probe for UGT1A1, showed sigmoidal kinetics with a Hill coefficient (n) of 1.2-1.3 in control and BNF-pretreated HepG2 cells. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 16951398-12 2006 It is suggested that the UGT1A1*28 genotype status could be used as a screening tool for a priori prevention of irinotecan-induced delayed-type diarrhea. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16791115-11 2006 Bilirubin increases were 2-fold higher in UGT1A1 [TA]7/[TA]7 genotypes. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16809730-0 2006 The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 16595709-10 2006 Despite limitations resulting from sample size, results indicate that UGT1A9 I399 and -118T(9/10) may represent additional candidates in combination with UGT1A1 promoter haplotypes for the prediction of SN-38 glucuronidation profile in vivo. Irinotecan 203-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-160 16595710-2 2006 Incubation of the DHT monoglucuronide with 12 cDNA-expressed recombinant human UGT isoforms and uridine 5"-diphosphoglucuronic acid resulted in a low but measurable DHT diglucuronidation activity primarily with UGT1A8, a gastrointestinal UGT, and to a lesser extent with UGT1A1 and UGT1A9. dht monoglucuronide 18-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 16595710-2 2006 Incubation of the DHT monoglucuronide with 12 cDNA-expressed recombinant human UGT isoforms and uridine 5"-diphosphoglucuronic acid resulted in a low but measurable DHT diglucuronidation activity primarily with UGT1A8, a gastrointestinal UGT, and to a lesser extent with UGT1A1 and UGT1A9. 5"-diphosphoglucuronic acid 104-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 16595710-2 2006 Incubation of the DHT monoglucuronide with 12 cDNA-expressed recombinant human UGT isoforms and uridine 5"-diphosphoglucuronic acid resulted in a low but measurable DHT diglucuronidation activity primarily with UGT1A8, a gastrointestinal UGT, and to a lesser extent with UGT1A1 and UGT1A9. Dihydrotestosterone 18-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 271-277 16824027-0 2006 Screening for adverse reactions to irinotecan treatment using the Invader UGT1A1 Molecular Assay. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-1 2006 Accumulating evidence provides support for the idea that determination of UGT1A1 polymorphisms before irinotecan (CPT-11) treatment is clinically useful and important for predicting and avoiding related toxicities. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-1 2006 Accumulating evidence provides support for the idea that determination of UGT1A1 polymorphisms before irinotecan (CPT-11) treatment is clinically useful and important for predicting and avoiding related toxicities. Irinotecan 114-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 16824027-3 2006 A dose reduction of irinotecan should be considered for patients known to be homozygous for the UGT1A1*28 allele when administered in combination with other agents or as a single agent. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 16824027-6 2006 This newly developed method for detecting UGT1A1 polymorphisms is feasible and has the potential to be widely used for rapid and accurate screening before irinotecan treatment. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16809730-1 2006 PURPOSE: UGT1A1*28 polymorphism has been associated with decreased glucuronidation of SN38, the active metabolite of irinotecan. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 16809730-11 2006 In particular, the possibility of a dose reduction for irinotecan in patients with a UGT1A1*28 polymorphism is not supported by the result of this analysis. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16783480-6 2006 The glucuronidation activity of niflumic acid in microsomes from eight human livers significantly correlated with UGT1A1-catalyzed estradiol 3beta-glucuronidation activity (r=0.78, p<0.05). Niflumic Acid 32-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 16783480-6 2006 The glucuronidation activity of niflumic acid in microsomes from eight human livers significantly correlated with UGT1A1-catalyzed estradiol 3beta-glucuronidation activity (r=0.78, p<0.05). Estradiol 131-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 16783480-8 2006 CONCLUSIONS: These findings indicate that UGT1A1 is the main isozyme involved in the glucuronidation of niflumic acid in the human liver. Niflumic Acid 104-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16713428-4 2006 Essentially, microsomal assays with UGT1A1 produced only one of the 2 N-glucuronides, whereas UGT1A9 is capable of forming both N-glucuronides. n-glucuronides 70-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 16504606-0 2006 Evaluation of mutation effects on UGT1A1 activity toward 17beta-estradiol using liquid chromatography-tandem mass spectrometry. Estradiol 57-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 16504606-1 2006 Mutations in the gene encoding UDP-glucuronosyltransferase 1A1 (UGT1A1) may reduce the glucuronidation of estradiol, bilirubin, etc. Estradiol 106-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-62 16504606-1 2006 Mutations in the gene encoding UDP-glucuronosyltransferase 1A1 (UGT1A1) may reduce the glucuronidation of estradiol, bilirubin, etc. Estradiol 106-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16504606-1 2006 Mutations in the gene encoding UDP-glucuronosyltransferase 1A1 (UGT1A1) may reduce the glucuronidation of estradiol, bilirubin, etc. Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-62 16504606-1 2006 Mutations in the gene encoding UDP-glucuronosyltransferase 1A1 (UGT1A1) may reduce the glucuronidation of estradiol, bilirubin, etc. Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16504606-2 2006 In the present study, we used a liquid chromatography-tandem mass spectrometry (LC/MS/MS) method to assay the activities of recombinant mutated UGT1A1 toward 17beta-estradiol (E2), by determining its glucuronide (E2G) content. Glucuronides 200-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 16508919-5 2006 Many enzymes are involved in controlling the disposition of irinotecan, including the cellular target (TOP1), metabolism enzymes (CES2, UGT1A1, CYP3A4, CYP3A5), and cellular transporters of the anti-cancer agent (ABCB1, ABCC1, ABCC2, ABCC3, ABCC5, ABCG2). Irinotecan 60-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 16713428-5 2006 The rates of metabolism for UGT1A9, human liver microsomes, and UGT1A1 were 200, 100, and 100 pmol N-glucuronide per minute per milligram of protein, respectively. n-glucuronide 99-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16685247-0 2006 Invader UGT1A1 molecular assay for irinotecan toxicity. Irinotecan 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 16817017-1 2006 During the course of the study of UGT1A1 induction by bilirubin, we could not detect the induction of the reporter gene (-3174/+14) of human UGT1A1 in HepG2 by bilirubin (Mol. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 16817017-4 2006 In this report, we show the finding of the induction of the reporter gene of UGT1A1 by cortisol at 1 microM, a major natural cortico-steroid, with human glucocorticoid receptor (GR). Hydrocortisone 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-83 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 94-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16636344-11 2006 CONCLUSION: These findings suggest that UGT1A1*6 and UGT1A9*22 genotypes may be important for SN-38 glucuronidation and associate with irinotecan-related severe toxicity. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16636344-12 2006 Specifically, UGT1A1*6 might be useful for predicting tumor response and survival outcome of Korean patients with NSCLC treated with irinotecan-based chemotherapy. Irinotecan 133-143 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 16682733-4 2006 For example, pharmacogenetic tests currently used in cancer therapy, such as genotyping UGT1A1 to reduce the incidence of severe toxicity of irinotecan and sequencing epidermal growth factor receptor from tumors to identify somatic mutations conferring sensitivity to tyrosine kinase inhibitors, were developed without initial identification of interpopulation differences. Irinotecan 141-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 16821585-4 2006 RESULTS: Three independent lines of enquiry indicated that, in the tumour specimens, SN-38 was glucuronidated primarily by UGT1A1, the isozyme generally recognised as being responsible for hepatic detoxification of this compound, while with NU/ICRF 505 two candidate isoforms emerged - UGT1A8 and/or UGT1A10 - both of which are not normally expressed in the liver. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 16628735-3 2006 UGT1A1 promoter polymorphism is associated both with unconjugated bilirubin level and elevated risk for cholelithiasis in such subset. Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16641252-4 2006 An assay is available for genotypic testing of the enzyme UGT1A1, which is predictive of toxicity from irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 16477579-6 2006 Among the UDP-glucuronosyltransferases (UGT) that are known to be expressed in the GI tract, the isoforms UGT1A1, 1A6, 1A8, 1A9 and 1A10 were active in glucuronidating trans- and/or cis-resveratrol. trans- and/ 168-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 16477579-6 2006 Among the UDP-glucuronosyltransferases (UGT) that are known to be expressed in the GI tract, the isoforms UGT1A1, 1A6, 1A8, 1A9 and 1A10 were active in glucuronidating trans- and/or cis-resveratrol. Resveratrol 182-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 16402080-1 2006 Retigabine (RGB) is an investigational antiepileptic drug, which undergoes extensive UGT1A1, 1A9 and 1A4-mediated N-glucuronidation and N-acetylation. ezogabine 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16402080-1 2006 Retigabine (RGB) is an investigational antiepileptic drug, which undergoes extensive UGT1A1, 1A9 and 1A4-mediated N-glucuronidation and N-acetylation. Nitrogen 114-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16513445-8 2006 Intestinal expression of UGT1A1, ABCB1, and ABCC2 was inversely correlated with the effects of ezetimibe on plant sterol serum concentrations. Sterols 114-120 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16715376-9 2006 Incubations with various cDNA-expressed UDP-glucuronosyltransferases indicated that the isozymes UGT1A1, UGT1A6, UGT1A8, and UGT1A9 were responsible for glucuronidation of 8-PN. 8-prenylnaringenin 172-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-103 16610035-1 2006 AIM: To identify the variants in UDP-glucuronosyltransferase 1 (UGT1A1) gene in Gilbert"s syndrome (GS) and to estimate the association between homozygosity for TA insertion and GS in India, as well as the frequency of TA insertion and its impact among normal controls in India. Tantalum 161-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16610035-1 2006 AIM: To identify the variants in UDP-glucuronosyltransferase 1 (UGT1A1) gene in Gilbert"s syndrome (GS) and to estimate the association between homozygosity for TA insertion and GS in India, as well as the frequency of TA insertion and its impact among normal controls in India. Tantalum 219-221 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16610035-5 2006 Functional assessment of a novel trinucleotide insertion was done by in silico analysis and by estimating UGT1A1 promoter activity carried out by luciferase reporter assay of appropriate constructs in Hep G2 cell line. trinucleotide 33-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 16597364-5 2006 UGT1A1 and UGT1A9 were predominantly responsible for the formation of the 3-O-glucuronide (Km = 149 microM) and 4"-O-glucuronide (Km = 365 microM), respectively. 3-o-glucuronide 74-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16597364-5 2006 UGT1A1 and UGT1A9 were predominantly responsible for the formation of the 3-O-glucuronide (Km = 149 microM) and 4"-O-glucuronide (Km = 365 microM), respectively. 4"-o-glucuronide 112-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16597364-7 2006 Resveratrol was co-incubated with substrates of UGT1A1 (bilirubin and 7-ethyl-10-hydroxycamptothecin (SN-38)) and UGT1A9 (7-hydroxytrifluoromethyl coumarin (7-HFC)). Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 16597364-10 2006 Hence, resveratrol has the potential to inhibit the glucuronidation of concomitantly administered therapeutic drugs or dietary components that are substrates of UGT1A1 and UGT1A9. Resveratrol 7-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 16456808-1 2006 BACKGROUND: In the current Phase II study, the authors evaluated the association between genomic polymorphic variants in uridine diphosphate glucuronosyl transferase (UGT1A1), methylenetetrahydrofolate reductase (MTHFR), and thymidylate synthase (TS) genes, and the incidence of the adverse effects of irinotecan and raltitrexed in previously heavily treated patients with metastatic colorectal carcinoma. Irinotecan 302-312 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 16456808-10 2006 CONCLUSIONS: In the current study, UGT1A1 promoter polymorphism was found to be predictive of the risk of diarrhea, emesis, and fatigue caused by chemotherapy with irinotecan and raltitrexed. Irinotecan 164-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 16456808-10 2006 CONCLUSIONS: In the current study, UGT1A1 promoter polymorphism was found to be predictive of the risk of diarrhea, emesis, and fatigue caused by chemotherapy with irinotecan and raltitrexed. raltitrexed 179-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 16456808-11 2006 Screening for UGT1A1 promoter polymorphism may be clinically useful for identifying patients at a higher risk of developing a severe or potentially life-threatening toxicity after irinotecan-based chemotherapy. Irinotecan 180-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Rifampin 83-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Glucuronides 174-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16513445-7 2006 RESULTS: Up-regulation of intestinal P-gp, MRP2, and UGT1A1 (but not of NPC1L1) by rifampin was associated with markedly decreased areas under the curve of ezetimibe and its glucuronide (116 +/- 78.1 ng.h/mL versus 49.9 +/- 31.0 ng.h/mL and 635 +/- 302 ng.h/mL versus 225 +/- 86.4 ng.h/mL, respectively; both P = .002) and increased intestinal clearances (2400 +/- 1560 mL/min versus 5500 +/- 4610 mL/min [P = .003] and 76.6 +/- 113 mL/min versus 316 +/- 457 mL/min [P = .010], respectively) and nearly abolished sterol-lowering effects. Sterols 513-519 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16513445-8 2006 Intestinal expression of UGT1A1, ABCB1, and ABCC2 was inversely correlated with the effects of ezetimibe on plant sterol serum concentrations. Ezetimibe 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16609363-0 2006 Relationship between hyperbilirubinaemia and UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism in adult HIV-infected Thai patients treated with indinavir. Indinavir 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-76 16609363-0 2006 Relationship between hyperbilirubinaemia and UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism in adult HIV-infected Thai patients treated with indinavir. Indinavir 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 16609363-8 2006 Total, conjugated (direct) and unconjugated (indirect) serum bilirubin concentrations increased significantly at 24 weeks of indinavir treatment for all four genotypes, with a trend towards higher levels depending on the number of UGT1A1 mutant alleles; *6/*28 > *6 > *28 > reference. Indinavir 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 231-237 16609363-9 2006 The hazards ratio (HR) for serious hyperbilirubinaemia (total bilirubin > 2.5 mg/dl) at week 24 was statistically significant only in those patients carrying the UGT1A1*6 (HR 2.87) and UGT1A1*6/*28 (HR 11.42) genotypes. Bilirubin 40-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 16609363-9 2006 The hazards ratio (HR) for serious hyperbilirubinaemia (total bilirubin > 2.5 mg/dl) at week 24 was statistically significant only in those patients carrying the UGT1A1*6 (HR 2.87) and UGT1A1*6/*28 (HR 11.42) genotypes. Bilirubin 40-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 188-194 16609363-10 2006 The Ki values for indinavir inhibition of UGT1A1 and UGT1A1*6 were 4.1 and 10.7 mumol/l respectively. Indinavir 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16609363-10 2006 The Ki values for indinavir inhibition of UGT1A1 and UGT1A1*6 were 4.1 and 10.7 mumol/l respectively. Indinavir 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16609363-13 2006 The Ki values determined for indinavir inhibition of UGT1A1 are consistent with an interaction in vivo, with an additive effect in patients with already impaired bilirubin glucuronidation activity. Indinavir 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16609363-13 2006 The Ki values determined for indinavir inhibition of UGT1A1 are consistent with an interaction in vivo, with an additive effect in patients with already impaired bilirubin glucuronidation activity. Bilirubin 162-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 16480962-6 2006 Only UGT1A4 catalyzed the N-linked glucuronidation of 4-HO-TAM among recombinant human UGT isoforms (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, UGT2B4, UGT2B7, UGT2B15, and UGT2B17) expressed in insect cells. 4-ho-tam 54-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 16610035-8 2006 Many new DNA variants in UGT1A1 gene were discovered, including a trinucleotide (CAT) insertion in the promoter found in a subset (10%) of GS patients, but not among normal controls. trinucleotide 66-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16610035-10 2006 CONCLUSION: The genetic epidemiology of GS is variable across ethnic groups and the epistatic interactions among UGT1A1 promoter variants modulate bilirubin glucuronidation. Bilirubin 147-156 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 16623861-0 2006 Neonatal jaundice and bilirubin UDP-glucuronosyl transferase 1A1 gene polymorphism in Turkish patients. Bilirubin 22-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-64 16623861-3 2006 The aim of this study was to investigate whether the number of thymine-adenine repeats in the promoter region of UGT-1A1 was related to non-physiologic hyperbilirubinemia of unexplained aetiology in Turkish newborns. thymine-adenine 63-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-120 16623861-4 2006 These patients (n=106) were genotyped for their thymine-adenine repeat number in the promoter region of UGT-1A1, and were divided into two groups according to their bilirubin level. thymine-adenine 48-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-111 16267566-5 2006 The absence of bilirubin UDP-glucoronyltransferase (UGT1A1) activity in CN patients causes high serum levels of unconjugated bilirubin and brain damage in infancy. Bilirubin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 16557566-4 2006 Our results show that patients with CF and gallstones are significantly more likely to carry at least one Gilbert UGT1A1 allele compared with stone-free patients (OR 7.3; P = .042) and that these carriers display significantly higher serum levels of unconjugated bilirubin (P = .002). Bilirubin 263-272 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 16551870-0 2006 Irinotecan inactivation is modulated by epigenetic silencing of UGT1A1 in colon cancer. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 16551870-2 2006 The UGT1A1-metabolizing enzyme, expressed in liver and colon, is primarily involved in the inactivation of its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 129-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 16551870-2 2006 The UGT1A1-metabolizing enzyme, expressed in liver and colon, is primarily involved in the inactivation of its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38). Irinotecan 161-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 16551870-5 2006 Bisulfite sequencing of the UGT1A1 gene revealed the aberrant methylation of specific CpG islands in UGT1A1-negative cells. hydrogen sulfite 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 16551870-5 2006 Bisulfite sequencing of the UGT1A1 gene revealed the aberrant methylation of specific CpG islands in UGT1A1-negative cells. hydrogen sulfite 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 16551870-9 2006 Loss of UGT1A1 methylation was further associated with an increase in UGT1A1 protein content and with an enhanced inactivation of SN-38 by 300% in HCT-116 cells. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 16551870-10 2006 CONCLUSIONS: We conclude that DNA methylation represses UGT1A1 expression in colon cancer and that this process may contribute to the level of tumoral inactivation of the anticancer agent SN-38 and potentially influence clinical response. Irinotecan 188-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 16513445-10 2006 CONCLUSIONS: The disposition and sterol-lowering effects of ezetimibe are modified by metabolic degradation of the drug via intestinal UGT1A1 and either intestinal or hepatic secretion (or both) via P-gp and MRP2. Ezetimibe 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 16877259-5 2006 Although the role of the UGT1A1 *28 genetic variant has received much attention in altered toxicity upon irinotecan treatment, other UGT1A enzymes also play an important role. Irinotecan 105-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16337205-3 2006 We have constructed a self-inactivating lentiviral vector (LV-ALBUGT) that expresses the human bilirubin UDP-glucuronosyltransferase (UGT1A1) from a liver-specific promoter. Bilirubin 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 16424820-0 2006 Study of the genetic determinants of UGT1A1 inducibility by phenobarbital in cultured human hepatocytes. Phenobarbital 60-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 16424820-1 2006 UGT1A1 is induced by phenobarbital. Phenobarbital 21-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16424820-6 2006 Significant induction of UGT1A1 catalytic activity was observed in 82% and 100% of the cultures treated with phenobarbital for 2 days (median fold-induction = 1.6, range 1.3-2.8; n = 28) and 6 days (median fold-induction = 2.8, range 1.6-6.4; n = 16), respectively. Phenobarbital 109-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 16288819-0 2006 Successful treatment of severe unconjugated hyperbilirubinemia via induction of UGT1A1 by rifampicin. Rifampin 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 17207411-4 2006 UGT1A1 activity has been measured in fluorescent assay using scopoletin as a substrate. Scopoletin 61-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16171463-0 2005 Role of cysteine residues in the function of human UDP glucuronosyltransferase isoform 1A1 (UGT1A1). Cysteine 8-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-90 16849011-1 2006 PURPOSE: It has been reported that carvedilol, which has beta-adrenergic blocking and vasodilating activities, is mainly metabolized by UDP-glucuronosyltransferase (UGT) 1A1, UGT2B4, UGT2B7 and CYP2D6. Carvedilol 35-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-173 16849011-8 2006 CONCLUSION: Polymorphisms of UGT1A1, UGT2B7 and CYP2D6 strongly affect the pharmacokinetics and disposition of carvedilol in Japanese. Carvedilol 111-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 16171463-7 2005 N-Ethylmaleimide did not inhibit UGT1A1 activity in native microsomes, but prevented UGT1A1 activation by UDP-GlcNAc and inhibited the activity in digitonin-permeabilized microsomes. Ethylmaleimide 0-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16171463-0 2005 Role of cysteine residues in the function of human UDP glucuronosyltransferase isoform 1A1 (UGT1A1). Cysteine 8-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 16171463-7 2005 N-Ethylmaleimide did not inhibit UGT1A1 activity in native microsomes, but prevented UGT1A1 activation by UDP-GlcNAc and inhibited the activity in digitonin-permeabilized microsomes. Uridine Diphosphate N-Acetylglucosamine 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16171463-1 2005 Bilirubin glucuronidation, catalysed by UGT1A1 [UGT (UDP glucuronosyltransferase) isoform 1A1, EC 2.4.1.17], is critical for biliary elimination of bilirubin. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16171463-9 2005 Together, the results suggested that free thiol groups, but not disulphide bonding, of seven cysteine residues within the intracisternal region of human UGT1A1 are important for its catalytic activity, while cysteine residues in the cytosolic domain may be involved in its physiological activation by UDP-GlcNAc. Sulfhydryl Compounds 42-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 16171463-1 2005 Bilirubin glucuronidation, catalysed by UGT1A1 [UGT (UDP glucuronosyltransferase) isoform 1A1, EC 2.4.1.17], is critical for biliary elimination of bilirubin. Bilirubin 148-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 40-46 16171463-9 2005 Together, the results suggested that free thiol groups, but not disulphide bonding, of seven cysteine residues within the intracisternal region of human UGT1A1 are important for its catalytic activity, while cysteine residues in the cytosolic domain may be involved in its physiological activation by UDP-GlcNAc. Cysteine 93-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 16171463-9 2005 Together, the results suggested that free thiol groups, but not disulphide bonding, of seven cysteine residues within the intracisternal region of human UGT1A1 are important for its catalytic activity, while cysteine residues in the cytosolic domain may be involved in its physiological activation by UDP-GlcNAc. Cysteine 208-216 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 16171463-3 2005 Nucleotide sequence analysis of UGT1A1 in two CN-1 patients revealed that patient A was homozygous for a nt 530 G-->A (where nt 530 G-->A means guanine to adenine transition at nucleotide 530) mutation, predicting a C177Y substitution, and patient B had a nt 466 T-->C mutation on one allele and a nt 1070 A-->G mutation on the other, predicting a C156R and a Q357R substitution respectively. Guanine 150-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 16171463-9 2005 Together, the results suggested that free thiol groups, but not disulphide bonding, of seven cysteine residues within the intracisternal region of human UGT1A1 are important for its catalytic activity, while cysteine residues in the cytosolic domain may be involved in its physiological activation by UDP-GlcNAc. Uridine Diphosphate N-Acetylglucosamine 301-311 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 16171463-3 2005 Nucleotide sequence analysis of UGT1A1 in two CN-1 patients revealed that patient A was homozygous for a nt 530 G-->A (where nt 530 G-->A means guanine to adenine transition at nucleotide 530) mutation, predicting a C177Y substitution, and patient B had a nt 466 T-->C mutation on one allele and a nt 1070 A-->G mutation on the other, predicting a C156R and a Q357R substitution respectively. Adenine 161-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 16171463-4 2005 All 11 cysteine residues of mature human UGT1A1 are highly conserved in other human UGT isoforms and in rat, mouse and Rhesus monkey UGT1A1, suggesting their functional importance. Cysteine 7-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 16171463-5 2005 Expression of mutagenized UGT1A1 plasmids showed that substitution of any of the seven cysteine residues located within the endoplasmic reticulum cisternae (including those mutated in patients A and B) abolished UGT1A1 activity or markedly increased its apparent K(m) for bilirubin. Cysteine 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16171463-5 2005 Expression of mutagenized UGT1A1 plasmids showed that substitution of any of the seven cysteine residues located within the endoplasmic reticulum cisternae (including those mutated in patients A and B) abolished UGT1A1 activity or markedly increased its apparent K(m) for bilirubin. Cysteine 87-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 16171463-5 2005 Expression of mutagenized UGT1A1 plasmids showed that substitution of any of the seven cysteine residues located within the endoplasmic reticulum cisternae (including those mutated in patients A and B) abolished UGT1A1 activity or markedly increased its apparent K(m) for bilirubin. Bilirubin 272-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16171463-5 2005 Expression of mutagenized UGT1A1 plasmids showed that substitution of any of the seven cysteine residues located within the endoplasmic reticulum cisternae (including those mutated in patients A and B) abolished UGT1A1 activity or markedly increased its apparent K(m) for bilirubin. Bilirubin 272-281 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 16171463-6 2005 Substitution of the three cysteine residues within the C-terminal cytosolic tail had minimal effect on basal UGT1A1 activity, but prevented UGT1A1 activation by UDP-GlcNAc. Cysteine 26-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 16171463-6 2005 Substitution of the three cysteine residues within the C-terminal cytosolic tail had minimal effect on basal UGT1A1 activity, but prevented UGT1A1 activation by UDP-GlcNAc. Uridine Diphosphate N-Acetylglucosamine 161-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 16491625-0 2005 UGT1A1 genotyping in patients undergoing treatment with irinotecan. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16051636-3 2005 It is known that different isoforms of UDP-glucuronosyltransferase (UGT) are involved in E2 glucuronidation: UGT1A1 leads exclusively to the 3-glucuronide and is stimulated by E2 via homotropic kinetics, whereas UGT2B7 gives rise to the 17-glucuronide of E2 following Michaelis-Menten kinetics. 3-glucuronide 141-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 16051636-3 2005 It is known that different isoforms of UDP-glucuronosyltransferase (UGT) are involved in E2 glucuronidation: UGT1A1 leads exclusively to the 3-glucuronide and is stimulated by E2 via homotropic kinetics, whereas UGT2B7 gives rise to the 17-glucuronide of E2 following Michaelis-Menten kinetics. 17-glucuronide 237-251 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 16051636-8 2005 The effect of daidzein on the 3-glucuronidation of E2 in human hepatic microsomes was also obtained with human recombinant UGT1A1. daidzein 14-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 16267624-0 2005 Pharmacogenetics of irinotecan: a promoter polymorphism of UGT1A1 gene and severe adverse reactions to irinotecan. Irinotecan 20-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 16267624-1 2005 This review focuses on a pharmacogenetic association between genetic polymorphism of UGT1A1 gene and severe adverse reactions to irinotecan. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 16267624-4 2005 A case-control study of Japanese cancer patients revealed that those with the variant UGT1A1 alleles were at significantly higher risk of severe adverse reactions to irinotecan, suggesting that the genotyping strategy would be clinically useful. Irinotecan 166-176 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 16187975-8 2005 UGT 1A1, 1A3, 1A6, 1A7, 1A8, 1A9, 1A10 and 2B7 glucuronidated naproxen. Naproxen 62-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-7 16118329-6 2005 By analysis of the double-reciprocal plots of bilirubin glucuronidation activities at different bilirubin concentrations in the presence of fixed concentrations of inhibitors, the UGT1A1 inhibition by atazanavir and indinavir was demonstrated to follow a linear mixed-type inhibition mechanism (Ki = 1.9 and 47.9 microM, respectively). Bilirubin 46-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 16118329-6 2005 By analysis of the double-reciprocal plots of bilirubin glucuronidation activities at different bilirubin concentrations in the presence of fixed concentrations of inhibitors, the UGT1A1 inhibition by atazanavir and indinavir was demonstrated to follow a linear mixed-type inhibition mechanism (Ki = 1.9 and 47.9 microM, respectively). Bilirubin 96-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 16118329-6 2005 By analysis of the double-reciprocal plots of bilirubin glucuronidation activities at different bilirubin concentrations in the presence of fixed concentrations of inhibitors, the UGT1A1 inhibition by atazanavir and indinavir was demonstrated to follow a linear mixed-type inhibition mechanism (Ki = 1.9 and 47.9 microM, respectively). Atazanavir Sulfate 201-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 180-186 16118329-7 2005 These results suggest that a direct inhibition of UGT1A1-mediated bilirubin glucuronidation may provide a mechanism for the reversible hyperbilirubinemia associated with administration of atazanavir as well as indinavir. Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 16118329-7 2005 These results suggest that a direct inhibition of UGT1A1-mediated bilirubin glucuronidation may provide a mechanism for the reversible hyperbilirubinemia associated with administration of atazanavir as well as indinavir. Atazanavir Sulfate 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 16118329-7 2005 These results suggest that a direct inhibition of UGT1A1-mediated bilirubin glucuronidation may provide a mechanism for the reversible hyperbilirubinemia associated with administration of atazanavir as well as indinavir. Indinavir 210-219 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 16019265-2 2005 A single injection of pDNA expressing hUGT1A1 under the CMV promoter resulted in excretion of bilirubin glucuronides in bile and a significant decrease in serum bilirubin for at least 2 or 4 weeks, respectively. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-45 16019265-2 2005 A single injection of pDNA expressing hUGT1A1 under the CMV promoter resulted in excretion of bilirubin glucuronides in bile and a significant decrease in serum bilirubin for at least 2 or 4 weeks, respectively. Glucuronides 104-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-45 16019265-2 2005 A single injection of pDNA expressing hUGT1A1 under the CMV promoter resulted in excretion of bilirubin glucuronides in bile and a significant decrease in serum bilirubin for at least 2 or 4 weeks, respectively. Bilirubin 161-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-45 16139098-4 2005 Further investigation identified multiple UGT isoforms to be responsible for catalyzing the addition of glucuronic acid to ethanol, with UGT1A1 and 2B7 being the two most prevalent isoforms. Glucuronic Acid 104-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 16139098-4 2005 Further investigation identified multiple UGT isoforms to be responsible for catalyzing the addition of glucuronic acid to ethanol, with UGT1A1 and 2B7 being the two most prevalent isoforms. Ethanol 123-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 16139098-5 2005 Co-incubation with bilirubin or 3"-azido-3"-deoxythymidine (UGT1A1 and 2B7 inhibitors, respectively) inhibited the greatest amount of ethyl glucuronide formation, though other UGT inhibitors also showed some effect. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-74 16139098-5 2005 Co-incubation with bilirubin or 3"-azido-3"-deoxythymidine (UGT1A1 and 2B7 inhibitors, respectively) inhibited the greatest amount of ethyl glucuronide formation, though other UGT inhibitors also showed some effect. Zidovudine 32-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-74 16139098-5 2005 Co-incubation with bilirubin or 3"-azido-3"-deoxythymidine (UGT1A1 and 2B7 inhibitors, respectively) inhibited the greatest amount of ethyl glucuronide formation, though other UGT inhibitors also showed some effect. ethyl glucuronide 134-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-74 16187975-11 2005 UGT 1A1, 1A7, 1A9 and 1A10 catalysed both the phenolic and acyl glucuronidation of desmethylnaproxen, while UGT 1A3, 1A6 and 2B7 formed only the acyl glucuronide. desmethylnaproxen 83-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-7 16187975-11 2005 UGT 1A1, 1A7, 1A9 and 1A10 catalysed both the phenolic and acyl glucuronidation of desmethylnaproxen, while UGT 1A3, 1A6 and 2B7 formed only the acyl glucuronide. acyl glucuronide 145-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-7 16166450-9 2005 Among the UGT1A isoforms screened, ketoconazole showed the highest inhibitory effect on UGT1A1 and UGT1A9. Ketoconazole 35-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 16237771-7 2005 The bilirubin levels for the cholelithiasis patients with the homozygous variant-UGT1A1 gene were significantly different from the control analog (18.0+/-6.5 and 12.7+/-2.9 micromol/L, respectively; P<0.001, Student"s t test). Bilirubin 4-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 16166450-11 2005 CONCLUSIONS: These results show that ketoconazole is a potent UGT1A1 inhibitor, which seems the basis for increased exposure to SN-38 when coadministered with irinotecan. Ketoconazole 37-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 16166450-11 2005 CONCLUSIONS: These results show that ketoconazole is a potent UGT1A1 inhibitor, which seems the basis for increased exposure to SN-38 when coadministered with irinotecan. Irinotecan 128-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 16166450-11 2005 CONCLUSIONS: These results show that ketoconazole is a potent UGT1A1 inhibitor, which seems the basis for increased exposure to SN-38 when coadministered with irinotecan. Irinotecan 159-169 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 15993909-9 2005 Genes in the conventional AH gene battery (e.g. CYP1A1, CYP1A2, CYP1B1, ALDH3A1, NQO1 and UGT1A1) remain responsive to TCDD in H/W rats despite the large deletion. Polychlorinated Dibenzodioxins 119-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 77-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 16187025-5 2005 The polymorphic UGT isoenzyme, UGT1A1 has genetic variants which decrease in SN-38 glucuronidating capacity and could help predict irinotecan-associated toxicity. Irinotecan 131-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 16187025-7 2005 We suggest that irinotecan offers an effective treatment option for children with recurrent brain stem glioma and other genetic variants except UGT1A1 may be a risk factor for irinotecan-induced toxicity. Irinotecan 176-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 15828025-5 2005 The main aim of this review is to provide an overview of the known polymorphisms present in the genes which codify for factors (thymidylate synthase dihydropyrimidine dehydrogenase, uridine diphosphate (UDP)-glucuronosyl-transferase 1A1, enzymes implicated in DNA repair) involved in the action mechanisms of the drugs now utilized in chemotherapeutic treatment of colorectal carcinoma, such as fluoropyrimidines, irinotecan, and platinum agents. fluoropyrimidines 395-412 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-236 15828025-5 2005 The main aim of this review is to provide an overview of the known polymorphisms present in the genes which codify for factors (thymidylate synthase dihydropyrimidine dehydrogenase, uridine diphosphate (UDP)-glucuronosyl-transferase 1A1, enzymes implicated in DNA repair) involved in the action mechanisms of the drugs now utilized in chemotherapeutic treatment of colorectal carcinoma, such as fluoropyrimidines, irinotecan, and platinum agents. Irinotecan 414-424 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 182-236 16092069-4 2005 The identification of the metabolites formed was elucidated using HPLC with diode array detection as well as HPLC/API-ES MS. XN was efficiently glucuronidated by UGT 1 A 8, 1 A 9, and 1 A 10; further important UGTs were UGT 1 A 1, 1 A 7, and 2 B 7. xanthohumol 125-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 220-229 16076256-0 2005 Single hepatic venous injection of liver-specific naked plasmid vector expressing human UGT1A1 leads to long-term correction of hyperbilirubinemia and prevention of chronic bilirubin toxicity in Gunn rats. Bilirubin 133-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15986396-8 2005 Using an heterologous expression system, the bilirubin-conjugating UGT1A1 enzyme was identified among all known UGTs (n = 16) as the predominant enzyme involved. Bilirubin 45-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 16004608-1 2005 PURPOSE: Homozygosity for the (AT)7 allele of uridine diphosphate glucuronosyl transferase 1A (UGT1A1) gene polymorphism is associated with increased bilirubin levels in sickle cell anemia (SCA). Bilirubin 150-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 16004608-13 2005 CONCLUSIONS: Apart from UGT1A1 (AT)7 homozygosity, Hb F, age and gender are the other factors that significantly influence serum bilirubin level in SCA. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 16083704-7 2005 RESULTS: RIFA enhanced bile acid detoxification as well as bilirubin conjugation and excretion as reflected by enhanced expression of CYP3A4, UGT1A1, and MRP2. Rifampin 9-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 142-148 16076256-3 2005 Although repeat injections of pcDNA3hUGT1A1 consistently reduced serum bilirubin levels, the effect did not exceed 2 weeks; hUGT1A1 was detectable in livers only for 2 weeks, despite the presence of vector and transcript for at least 1 month. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-43 16012956-2 2005 Gilbert"s syndrome is recessively inherited, unconjugated hyperbilirubinemia caused by decreased conjugation rate of bilirubin associated mostly with homozygous A(TA) 7 TAA variant of the TATAA-box in the UGT1A1 gene promoter. Bilirubin 63-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 15921999-2 2005 Three substrates were chosen to cover both UGT1A and UGT2B family isozymes: bilirubin (substrate of UGT1A1), p-nitrophenol (UGT1A6) and ethinylestradiol (UGT2B1 and 2B3 for position C17 and UGT1A1 for position C3). Bilirubin 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 15930350-8 2005 Patients with UDP-glucuronosyltransferase (UGT1A1; TA)6/6 promoter genotype had a lower ratio of free to glucuronide form of SN-38 than in patients with >/=1 (TA)7 allele. Glucuronides 105-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 15896333-2 2005 In this study, the effects of sulforaphane (SFN) and its sulfide analog, erucin (ERN), have been examined on the induction of the phase II enzymes, quinine oxidoreductase (NQO1) and UDP-glucuronosyl transferase (UGT1A1), multidrug transporter (MRP2), cell cycle arrest and cell death in human colon adenocarcinoma Caco-2 cells. sulforaphane 30-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 15896333-2 2005 In this study, the effects of sulforaphane (SFN) and its sulfide analog, erucin (ERN), have been examined on the induction of the phase II enzymes, quinine oxidoreductase (NQO1) and UDP-glucuronosyl transferase (UGT1A1), multidrug transporter (MRP2), cell cycle arrest and cell death in human colon adenocarcinoma Caco-2 cells. sulforaphane 44-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 15896333-5 2005 Erucin (20 microM) induced the mRNAs of NQO1, UGT1A1 and MRP2 by 11.1-, 11.6- and 6.7-fold, whereas sulforaphane (20 microM) induced 3.3-, 5.3- and 2.2-fold, respectively. erucin 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 15930350-8 2005 Patients with UDP-glucuronosyltransferase (UGT1A1; TA)6/6 promoter genotype had a lower ratio of free to glucuronide form of SN-38 than in patients with >/=1 (TA)7 allele. Irinotecan 125-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 15716465-11 2005 Its association with variants of the UGT1A1 promoter and UGT1A7 gene may influence irinotecan metabolism. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 15769885-0 2005 Acyl glucuronidation of fluoroquinolone antibiotics by the UDP-glucuronosyltransferase 1A subfamily in human liver microsomes. Fluoroquinolones 24-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-89 15769885-5 2005 In a bank of human liver microsomes (n = 14), the glucuronidation activities of LVFX, MFLX, and STFX correlated highly with UGT1A1-selective beta-estradiol 3-glucuronidation activity, whereas the glucuronidation activity of GPFX correlated highly with UGT1A9-selective propofol glucuronidation activity. Moxifloxacin 86-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 15769885-5 2005 In a bank of human liver microsomes (n = 14), the glucuronidation activities of LVFX, MFLX, and STFX correlated highly with UGT1A1-selective beta-estradiol 3-glucuronidation activity, whereas the glucuronidation activity of GPFX correlated highly with UGT1A9-selective propofol glucuronidation activity. sitafloxacin 96-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 15769885-5 2005 In a bank of human liver microsomes (n = 14), the glucuronidation activities of LVFX, MFLX, and STFX correlated highly with UGT1A1-selective beta-estradiol 3-glucuronidation activity, whereas the glucuronidation activity of GPFX correlated highly with UGT1A9-selective propofol glucuronidation activity. Propofol 269-277 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 15769885-6 2005 Among 12 recombinant UGT enzymes, UGT1A1, 1A3, 1A7, and 1A9 catalyzed the glucuronidation of these fluoroquinolones. Fluoroquinolones 99-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 15858133-3 2005 A heterozygous G > A transition at the 5" splicing donor consensus sequence in intron 14 leading to exon 14 skipping (IVS14+1 G > A, DPYD*2A) with partial loss of enzyme activity may be partly responsible for 5FU induced toxicity, whereas irinotecan associated toxicity may in part be explained by an aberrant UGT1A1 promoter (TA)(n) genotype underlying Gilbert"s syndrome with reduced liver glucuronidation activity. Fluorouracil 215-218 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 316-322 15858133-3 2005 A heterozygous G > A transition at the 5" splicing donor consensus sequence in intron 14 leading to exon 14 skipping (IVS14+1 G > A, DPYD*2A) with partial loss of enzyme activity may be partly responsible for 5FU induced toxicity, whereas irinotecan associated toxicity may in part be explained by an aberrant UGT1A1 promoter (TA)(n) genotype underlying Gilbert"s syndrome with reduced liver glucuronidation activity. Irinotecan 245-255 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 316-322 15953334-2 2005 A Chinese girl with severe jaundice was recently diagnosed to have CN syndrome type I by analyzing the bilirubin-uridinediphospho (UDP)-glucuronosyltransferase gene (UGT1A1). Bilirubin 103-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 15716465-2 2005 UGT1A1*28 is a functional UGT promoter polymorphism associated with Gilbert"s disease and severe irinotecan toxicity, which also occurs in the absence of UGT1A1*28. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15988124-4 2005 The TA-1801A glucuronosyltransferase activity in human liver microsomes was inhibited by bilirubin (typical substrate for UGT1A1), propofol (typical substrate for UGT1A9), diclofenac (substrate for UGT1A9 and UGT2B7), and genistein (substrate for UGT1A1, UGT1A3, and UGT1A9). Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 15988124-4 2005 The TA-1801A glucuronosyltransferase activity in human liver microsomes was inhibited by bilirubin (typical substrate for UGT1A1), propofol (typical substrate for UGT1A9), diclofenac (substrate for UGT1A9 and UGT2B7), and genistein (substrate for UGT1A1, UGT1A3, and UGT1A9). Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 247-253 15988124-4 2005 The TA-1801A glucuronosyltransferase activity in human liver microsomes was inhibited by bilirubin (typical substrate for UGT1A1), propofol (typical substrate for UGT1A9), diclofenac (substrate for UGT1A9 and UGT2B7), and genistein (substrate for UGT1A1, UGT1A3, and UGT1A9). Propofol 131-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 247-253 15988124-5 2005 The inhibition by bilirubin, propofol, and diclofenac of the TA-1801A glucuronidation was less pronounced in jejunum microsomes than liver microsomes, suggesting that the contribution of UGT1A1, UGT1A9, and UGT2B7 to the TA-1801A glucuronidation is smaller in the intestine than the liver. Bilirubin 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 15988124-5 2005 The inhibition by bilirubin, propofol, and diclofenac of the TA-1801A glucuronidation was less pronounced in jejunum microsomes than liver microsomes, suggesting that the contribution of UGT1A1, UGT1A9, and UGT2B7 to the TA-1801A glucuronidation is smaller in the intestine than the liver. Propofol 29-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 15988124-5 2005 The inhibition by bilirubin, propofol, and diclofenac of the TA-1801A glucuronidation was less pronounced in jejunum microsomes than liver microsomes, suggesting that the contribution of UGT1A1, UGT1A9, and UGT2B7 to the TA-1801A glucuronidation is smaller in the intestine than the liver. Diclofenac 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-193 15867280-0 2005 Cruciferae interact with the UGT1A1*28 polymorphism to determine serum bilirubin levels in humans. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 15867280-1 2005 UDP-glucuronosyltransferase (UGT) 1A1 is a conjugating biotransformation enzyme that plays a role in maintaining levels of endogenous compounds (e.g., bilirubin) and handling exogenous compounds, including carcinogens. Bilirubin 151-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 15867280-2 2005 The UGT1A1*28 polymorphism results in decreased UGT1A1 promoter activity due to 7 thymine-adenine (TA) repeats instead of the commonly found 6 repeats. 7 thymine-adenine 80-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 15867280-2 2005 The UGT1A1*28 polymorphism results in decreased UGT1A1 promoter activity due to 7 thymine-adenine (TA) repeats instead of the commonly found 6 repeats. 7 thymine-adenine 80-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15867280-2 2005 The UGT1A1*28 polymorphism results in decreased UGT1A1 promoter activity due to 7 thymine-adenine (TA) repeats instead of the commonly found 6 repeats. Tantalum 99-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 15867280-2 2005 The UGT1A1*28 polymorphism results in decreased UGT1A1 promoter activity due to 7 thymine-adenine (TA) repeats instead of the commonly found 6 repeats. Tantalum 99-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15867280-8 2005 There was a significant inverse association between all 3 bilirubin measures and interaction of UGT1A1*28 genotype with Cruciferae intake (P < 0.02 for each measure); individuals with the 7/7 genotype had reduced bilirubin concentrations with increased intake of cruciferous vegetables, whereas individuals with the 6/6 or 6/7 genotype did not. Bilirubin 216-225 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 15867280-9 2005 With regard to UGT1A1-conjugated carcinogens (e.g., heterocyclic amines, polycyclic aromatic hydrocarbons), individuals with decreased UGT1A1 activity due to the 7/7 genotype may be at greater risk for carcinogenesis, but our results imply that they also may have greater opportunity to decrease that risk through dietary intervention. heterocyclic amines 52-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 15867280-9 2005 With regard to UGT1A1-conjugated carcinogens (e.g., heterocyclic amines, polycyclic aromatic hydrocarbons), individuals with decreased UGT1A1 activity due to the 7/7 genotype may be at greater risk for carcinogenesis, but our results imply that they also may have greater opportunity to decrease that risk through dietary intervention. Polycyclic Aromatic Hydrocarbons 73-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 15855726-2 2005 Previously, we demonstrated that the glucuronidation of 4"-HPPH is catalyzed by multiple UDP-glucuronosyltransferases (UGTs) of UGT1A1, UGT1A4, UGT1A6, and UGT1A9. hydroxyphenytoin 56-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 128-134 15801936-3 2005 All patients were genotyped for allelic variants in genes encoding drug metabolizing enzymes (CYP3A4, CYP3A5, UGT1A1) and drug transporters (ABCB1, ABCC2 and ABCG2) that are involved in irinotecan disposition. Irinotecan 186-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15864130-2 2005 SN-38, the active metabolite of the anticancer agent irinotecan, is metabolized by both UGT1A1 and UGT1A9. Irinotecan 53-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15853761-11 2005 Since UGT1A1 metabolizes not only bilirubin but also hormones and drugs, the mutations could be involved in carcinogenesis and adverse drug reactions. Bilirubin 34-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 15855726-4 2005 In the present study, we investigated the relationship between the extent of interindividual variability in the urinary excretion levels of 4"-HPPH and its O-glucuronide and genotyping of CYP2C9, CYP2C19, UGT1A1, UGT1A6, and UGT1A9. hydroxyphenytoin 140-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 15777743-1 2005 UDP-glucuronosyltransferase1A1 (UGT1A1) catalyses glucuronidation of bilirubin (the final break down product of heme which is produced mainly in the spleen and liver) and is located on the lumen of the endoplasmic reticulum (ER). Bilirubin 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 15777743-1 2005 UDP-glucuronosyltransferase1A1 (UGT1A1) catalyses glucuronidation of bilirubin (the final break down product of heme which is produced mainly in the spleen and liver) and is located on the lumen of the endoplasmic reticulum (ER). Bilirubin 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 15777743-1 2005 UDP-glucuronosyltransferase1A1 (UGT1A1) catalyses glucuronidation of bilirubin (the final break down product of heme which is produced mainly in the spleen and liver) and is located on the lumen of the endoplasmic reticulum (ER). Heme 112-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 15777743-1 2005 UDP-glucuronosyltransferase1A1 (UGT1A1) catalyses glucuronidation of bilirubin (the final break down product of heme which is produced mainly in the spleen and liver) and is located on the lumen of the endoplasmic reticulum (ER). Heme 112-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 15949398-9 2005 The levels of UGT1A1, UGT1A8, and UGT1A10 mRNA expression were significantly increased in the cells treated with 25 micromol/L sulforaphane compared to that in the controls (P = 0.006, P = 0.017, and P = 0.008 respectively). sulforaphane 127-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 15985997-5 2005 Mutations in the gene encoding bilirubin-UGT (UGT1A1), lead to complete or partial inactivation of the enzyme causing the rare autosomal recessively inherited conditions, Crigler-Najjar syndrome type 1 (CN-1) and type 2 (CN-2). Bilirubin 31-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 15771689-3 2005 Recently, it was reported that the -3263T > G mutation in the phenobarbital response enhancer module in UGT1A1 was associated with the majority of cases of Gilbert syndrome. Phenobarbital 65-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 15949398-14 2005 CONCLUSION: (1) Low dose sulforaphane induces the expression of UGT1A, UGT1A1, UGT1A A8, and UGT1A A10 mRNA significantly. sulforaphane 25-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 15572581-10 2005 The intrinsic clearance of 7-ethyl-10-hydroxycamptothecin (SN-38) by P229L UGT1A1 expressed in COS-1 cells was about 3% of the wild type. Irinotecan 27-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 15572581-10 2005 The intrinsic clearance of 7-ethyl-10-hydroxycamptothecin (SN-38) by P229L UGT1A1 expressed in COS-1 cells was about 3% of the wild type. Irinotecan 59-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 15572581-10 2005 The intrinsic clearance of 7-ethyl-10-hydroxycamptothecin (SN-38) by P229L UGT1A1 expressed in COS-1 cells was about 3% of the wild type. carbonyl sulfide 95-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-81 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-203 15710570-2 2005 We investigated whether susceptibility to cholelithiasis in SCA was associated with the promoter polymorphism of the 5?-diphosphate-glucuronosyltransferase 1A1 (UGT1A1) gene encoding a key enzyme in bilirubin catabolism. Bilirubin 199-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 161-167 15557560-1 2005 UDP-glucuronosyltransferase (UGT) 1A1 glucuronidates endogenous metabolites, such as bilirubin, and exogenous substances, and plays a critical role in their detoxification and excretion. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 15557560-2 2005 In a previous article, we described the phenobarbital response activity to a 290-base pair (bp) distal enhancer sequence (-3499/-3210) of the human UGT1A1 gene that is activated by the constitutive androstane receptor (CAR). Phenobarbital 40-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 148-154 15557560-3 2005 Here, we show that dexamethasone at submicromolar concentrations enhances the pregnane X receptor (PXR) activator-mediated expression of the UGT1A1 gene and protein in HepG2 cells. Dexamethasone 19-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-147 15727486-10 2005 The enzymes responsible for the activation, metabolism and mechanism of action of irinotecan, namely carboxylesterase 2, cytochrome P450 (CYP) 3A4, uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1), and topoisomerase-I, also exhibit variable interindividual activity. Irinotecan 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 205-211 15727486-13 2005 CYP3A4 and UGT1A1 activity levels might be predictive of irinotecan toxicity rather than efficacy. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 15881424-1 2005 Crigler-Najjar syndrome type 1 (CN1) is an inherited disorder characterized by the absence of hepatic uridine diphosphoglucuronate glucuronosyltransferase (UDPGT), the enzyme responsible for the conjugation and excretion of bilirubin. Bilirubin 224-233 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 156-161 15593333-0 2005 In vitro inhibitory effects of non-steroidal antiinflammatory drugs on UDP-glucuronosyltransferase 1A1-catalysed estradiol 3beta-glucuronidation in human liver microsomes. Estradiol 113-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-102 15593333-6 2005 The IC50 value of diclofenac for 4-methylumbelliferone glucuronidation in recombinant human UGT1A1 was 57.5 microM, similar to that obtained for E3G using HLM. Diclofenac 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 15593333-6 2005 The IC50 value of diclofenac for 4-methylumbelliferone glucuronidation in recombinant human UGT1A1 was 57.5 microM, similar to that obtained for E3G using HLM. Hymecromone 33-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 15593333-7 2005 In conclusion, niflumic acid had the most potent inhibitory effects on UGT1A1-catalysed E3G in HLM among seven NSAID investigated. Niflumic Acid 15-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 15881424-5 2005 After 2 weeks phenobarbitone was added to promote the enzymatic activity of UDPGT of the transplanted hepatocytes. Phenobarbital 14-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-81 15720263-8 2005 Uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) genotype has been reported to be associated with time to progression and survival in patients treated with irinotecan. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-55 15720263-8 2005 Uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1) genotype has been reported to be associated with time to progression and survival in patients treated with irinotecan. Irinotecan 172-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 16399340-6 2005 In humans, lethal hyperbilirubinemic Crigler-Najjar type 1 and milder diseases/syndromes are due to deleterious to mildly deleterious mutations in the bilirubin-specific UGT1A1 or a common exon. Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-176 16257834-2 2005 Irinotecan undergoes drug metabolism to form an active SN-38, which is further converted to its beta-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-153 16257834-2 2005 Irinotecan undergoes drug metabolism to form an active SN-38, which is further converted to its beta-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 55-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-153 16257834-2 2005 Irinotecan undergoes drug metabolism to form an active SN-38, which is further converted to its beta-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. beta-glucuronide 96-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-153 16257834-3 2005 A variant in the promoter of UGT1A1 gene, UGT1A1*28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and ADR to irinotecan have been reported. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 16257834-3 2005 A variant in the promoter of UGT1A1 gene, UGT1A1*28 allele, has been extensively studied, and pharmacogenetic relationships between the variant and ADR to irinotecan have been reported. Irinotecan 155-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 16257834-4 2005 A case-control study of Japanese cancer patients demonstrated that the patients having UGT1A1*28 were at significantly increased risk of severe ADR to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 87-93 16257834-5 2005 To date, genetic variations of the UGT1A1 gene is the most important hereditary factor to predict severe ADR to irinotecan. Irinotecan 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 16257834-7 2005 At present, irinotecan chemotherapy based on a patient"s UGT1A1 genetic status is scientifically reasonable. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 15849716-3 2005 Concentration-dependent changes in UGT1A1 response were evaluated in hepatocyte cultures after treatment with 3-methylchloranthrene, beta-napthoflavone, rifampicin, or phenobarbital. 3-methylchloranthrene 110-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 15849716-3 2005 Concentration-dependent changes in UGT1A1 response were evaluated in hepatocyte cultures after treatment with 3-methylchloranthrene, beta-napthoflavone, rifampicin, or phenobarbital. beta-napthoflavone 133-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 15849716-3 2005 Concentration-dependent changes in UGT1A1 response were evaluated in hepatocyte cultures after treatment with 3-methylchloranthrene, beta-napthoflavone, rifampicin, or phenobarbital. Rifampin 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 15849716-3 2005 Concentration-dependent changes in UGT1A1 response were evaluated in hepatocyte cultures after treatment with 3-methylchloranthrene, beta-napthoflavone, rifampicin, or phenobarbital. Phenobarbital 168-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 15849716-6 2005 Pharmacodynamic modeling revealed EC50 values statistically significant between UGT1A1 and CYP2B6 after treatment with PB, but not statistically distinguishable between UGT1A1 and CYP"s 1A2 or 3A4 after treatment with 3-methylchloranthrene or rifampicin, respectively. Lead 119-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Rifampin 63-73 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Ritonavir 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Clotrimazole 90-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Phenobarbital 195-208 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15849716-7 2005 UGT1A1 was most responsive to the pregnane-X-receptor-agonists rifampicin, ritonavir, and clotrimazole at the mRNA level and, to a lesser extent, the constitutive androstane receptor-activators, phenobarbital and phenytoin. Phenytoin 213-222 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 16399344-8 2005 Similar to regulation of CYP1A1, the transcriptional activation of UGT1A1 by TCDD is mediated through the aryl hydrocarbon receptor. Polychlorinated Dibenzodioxins 77-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 16399345-1 2005 Human UDP-glucuronosyltransferase (UGT) 1A1 is the enzyme that detoxifies neurotoxic bilirubin by conjugating it with glucuronic acid. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 16399345-1 2005 Human UDP-glucuronosyltransferase (UGT) 1A1 is the enzyme that detoxifies neurotoxic bilirubin by conjugating it with glucuronic acid. Glucuronic Acid 118-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 16399345-2 2005 In addition to bilirubin, UGT1A1 conjugates various endogenous and exogenous lipophilic compounds such as estrogens and the active metabolite of the anticancer drug irinotecan SN-38. Irinotecan 165-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16399345-2 2005 In addition to bilirubin, UGT1A1 conjugates various endogenous and exogenous lipophilic compounds such as estrogens and the active metabolite of the anticancer drug irinotecan SN-38. Irinotecan 176-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 16399345-3 2005 Thus, activation by specific inducers of the UGT1A1 gene is critical in treating patients with unconjugated hyperbili-rubinemia and in preventing side effects of drug treatment such as SN-38-induced toxicity. Irinotecan 185-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 15965581-1 2005 In bilirubin metabolism, increased destruction of erythrocytes, defect in the function of organic anion transporter polypeptide 2 (OATP2) or UDP-glucuronosyltransferase 1A1 (UGT1A1) may result in unconjugated hyperbilirubinemia. Bilirubin 3-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 141-172 15965581-5 2005 The status of the haplotypes of G6PD, OATP2, and UGT1A1 genes affected the odds ratio and the bilirubin levels in the hyperbilirubinemic subjects. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 16399344-5 2005 UDP-glucuronosyltransferase 1A1 (UGT1A1), responsible for the glucuronidation of bilirubin, is controlled in a tissue-specific manner and can be regulated following environmental exposure. Bilirubin 81-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 16399344-5 2005 UDP-glucuronosyltransferase 1A1 (UGT1A1), responsible for the glucuronidation of bilirubin, is controlled in a tissue-specific manner and can be regulated following environmental exposure. Bilirubin 81-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 16399344-6 2005 This chapter describes materials and methods for the examination of molecular interactions that control UGT1A1 expression and induction in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Polychlorinated Dibenzodioxins 151-186 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 16399344-6 2005 This chapter describes materials and methods for the examination of molecular interactions that control UGT1A1 expression and induction in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Polychlorinated Dibenzodioxins 188-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 15378558-1 2004 The effects of beta-estradiol and propofol on human UGT1A1, 1A8 and 1A9 activities were investigated using 4-methylumbelliferone (4-MU) as a substrate for glucuronidation. Estradiol 15-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 15724447-4 2005 The activities of detoxifying enzymes such as glutathione transferase (GST), quinone reductase (QR) and UDP-glucuronosyl transferase (UDP-GT) were found to be decreased while the hydrogen peroxide level was increased in the lung cancer bearing animals. Hydrogen Peroxide 179-196 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 134-140 15864124-0 2005 Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Phenobarbital 28-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-107 15864124-0 2005 Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Irinotecan 117-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-107 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 168-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 231-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-65 15864124-1 2005 Genetic polymorphism of the UDP-glucuronosyltransferase (UGT) 1A1 gene is associated with the decreased glucuronidation activity of an active metabolite of irinotecan, SN-38, and UGT1A1*28 has been shown as a predictive factor for irinotecan toxicity. Irinotecan 231-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 15864124-2 2005 The phenobarbital-responsive enhancer module (PBREM) of the UGT1A1 promoter region has been reportedly associated with the transcriptional activity of the gene. Phenobarbital 4-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 15864124-12 2005 The determination of T-3279G and UGT1A1*28 genotypes might be clinically useful in predicting severe irinotecan toxicity in cancer patients. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15684703-2 2004 After delivery of pDNA encoding hUGT1A1 via hepatic vein or femoral artery, in vitro bilirubin glucuronidation activity was detectable in Gunn rat liver and muscle extracts. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-39 15684703-3 2004 Expression of hUGT1A1 in Gunn rat liver or muscle resulted in excretion of bilirubin glucuronides in bile. Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-21 15684703-3 2004 Expression of hUGT1A1 in Gunn rat liver or muscle resulted in excretion of bilirubin glucuronides in bile. Glucuronides 85-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-21 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-237 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 16-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 239-245 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-237 15523087-2 2004 The response to irinotecan is variable, possibly because of interindividual variation in the expression of the enzymes that metabolize irinotecan, including cytochrome P450 3A4 (CYP3A4) and uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1). Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 239-245 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 15523087-12 2004 In addition, the presence of a UGT1A1 variant with a (TA)7 repeat in the promoter (UGT1A1*28) was associated with increased exposure to SN-38 (435 ng x h/mL, 95% confidence interval [CI] = 339 to 531 ng x h/mL in patients who are homozygous for wild-type UGT1A1; 631 ng x h/mL, 95% CI = 499 to 762 ng . Irinotecan 136-141 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 15523087-15 2004 Evaluation of midazolam clearance combined with UGT1A1*28 genotyping may assist with optimization of irinotecan chemotherapy. Irinotecan 101-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15320866-0 2004 Farnesol is glucuronidated in human liver, kidney and intestine in vitro, and is a novel substrate for UGT2B7 and UGT1A1. Farnesol 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 15320866-8 2004 Kinetic analysis and inhibition experiments indicate that, in liver microsomes, UGT1A1 is primarily responsible for farnesol glucuronidation; however, in intestine microsomes, UGT2B7 is probably the major isoform involved, with a very-low-micromolar K(m). Farnesol 116-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 15378558-1 2004 The effects of beta-estradiol and propofol on human UGT1A1, 1A8 and 1A9 activities were investigated using 4-methylumbelliferone (4-MU) as a substrate for glucuronidation. Propofol 34-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 15378558-1 2004 The effects of beta-estradiol and propofol on human UGT1A1, 1A8 and 1A9 activities were investigated using 4-methylumbelliferone (4-MU) as a substrate for glucuronidation. Hymecromone 107-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 15378558-2 2004 The formation of 4-MU glucuronide (4-MUG) from 4-MU, in recombinant human UGT 1A1, 1A8 and 1A9 was determined using HPLC with fluorescence detection. 4-methylumbelliferyl glucuronide 17-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-81 15378558-4 2004 beta-estradiol had potent inhibitory effects on UGT1A9 as well as on UGT1A1 with IC(50) values of 2.1 and 7.2 microM, respectively. Estradiol 0-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 15378558-6 2004 In contrast, beta-estradiol and propofol activated 4-MU glucuronidation in UGT1A1 and 1A8, respectively. Estradiol 13-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-89 15378558-6 2004 In contrast, beta-estradiol and propofol activated 4-MU glucuronidation in UGT1A1 and 1A8, respectively. Propofol 32-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 75-89 15378558-7 2004 This study therefore indicates that the use of beta-estradiol as a specific inhibitor for UGT1A1 should be used with care in the identification of UGT isozymes responsible for glucuronidation in human liver microsomes. Estradiol 47-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 15688606-3 2004 In particular, uridine diphosphoglucuronosyl transferase 1A1 (UGT1A1) enzyme is responsible for detoxification of irinotecan active metabolite, SN38. Irinotecan 114-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 15688606-4 2004 A polymorphic structure in the promoter region (UGT1A1*28) may affect irinotecan toxicity and SN38 plasma level. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 15269186-8 2004 In contrast, tocotrienols induce MDR1 and UGT1A1 but not CYP3A4 expression in intestinal LS180 cells. Tocotrienols 13-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 15519273-0 2004 Influence of common variants in the pharmacokinetic genes (OATP-C, UGT1A1, and MRP2) on serum bilirubin levels in healthy subjects. Bilirubin 94-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Phenobarbital 61-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 15334623-6 2004 Inhibition studies with known inhibitors of UGT (diclofenac, flunitrazepam and bilirubin) confirmed the involvement of UGT1A1, UGT1A3 and UGT2B15 in the formation of 3-hydroxydesloratadine-glucuronide. 3-hydroxydesloratadine-glucuronide 166-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 15469406-3 2004 Although multiple genes may play a role in irinotecan activity, the UDP glycuronosyltransferase 1 family, polypeptide A1 (UGT1A1) enzyme has been strongly associated with toxicity. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Dinucleoside Phosphates 9-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Dinucleoside Phosphates 9-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Irinotecan 154-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 15469406-4 2004 A common dinucleotide repeat polymorphism in the UGT1A1 promoter region (UGT1A1*28) has been correlated with severe toxicity in cancer patients receiving irinotecan-containing therapy. Irinotecan 154-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 15517893-5 2004 RESULTS: Our HPLC assay results showed that both UGT1A1 and UGT1A10 are responsible for SN-38 glucuronidation. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 49-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 15517893-6 2004 The levels of UGT1A1 and UGT1A10 expression in a CPT-11/SN-38-resistant cell line were increased compared to levels in the parent cell line. Irinotecan 56-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 14-20 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 101-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 108-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 140-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15517893-9 2004 Our results suggest that not only UGT 1A1, but also UGT 1A10, plays an important role in detoxifying CPT-11/SN-38, leading to resistance to CPT-11/SN-38 in lung cancer. Irinotecan 147-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 15334623-5 2004 Out of ten recombinant human UDP-glucuronosyltransferases (UGTs), UGT1A1, UGT1A3, UGT1A8 and UGT2B15 exhibited catalytic activity with respect to the formation of 3-hydroxydesloratadine-glucuronide. 3-hydroxydesloratadine-glucuronide 163-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 15334623-6 2004 Inhibition studies with known inhibitors of UGT (diclofenac, flunitrazepam and bilirubin) confirmed the involvement of UGT1A1, UGT1A3 and UGT2B15 in the formation of 3-hydroxydesloratadine-glucuronide. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 119-125 15334623-7 2004 The results from this study demonstrated that the in vitro formation of 3-hydroxydesloratadine-glucuronide from 3-hydroxydesloratadine was mediated via UGT1A1, UGT1A3 and UGT2B15 in human liver. 3-hydroxydesloratadine-glucuronide 72-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 15334623-7 2004 The results from this study demonstrated that the in vitro formation of 3-hydroxydesloratadine-glucuronide from 3-hydroxydesloratadine was mediated via UGT1A1, UGT1A3 and UGT2B15 in human liver. 3-hydroxydesloratadine 72-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 152-158 15560369-0 2004 Stimulation of transcriptional expression of human UDP-glucuronosyltransferase 1A1 by dexamethasone. Dexamethasone 86-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 51-82 15090468-0 2004 Interactions between sulforaphane and apigenin in the induction of UGT1A1 and GSTA1 in CaCo-2 cells. sulforaphane 21-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 15090468-0 2004 Interactions between sulforaphane and apigenin in the induction of UGT1A1 and GSTA1 in CaCo-2 cells. Apigenin 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 15090468-3 2004 We show that apigenin induces UGT1A1 transcription (4-fold) but not GSTA1, and that sulforaphane induces both UGT1A1 (3.7-fold) and GSTA1 (2.8-fold) transcription in both dose- and time-dependent manners. sulforaphane 84-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 15090468-4 2004 The combination of sulforaphane and apigenin resulted in a synergistic induction of UGT1A1 mRNA up to 12-fold, although this interaction was not seen for GSTA1. sulforaphane 19-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 15090468-4 2004 The combination of sulforaphane and apigenin resulted in a synergistic induction of UGT1A1 mRNA up to 12-fold, although this interaction was not seen for GSTA1. Apigenin 36-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. sulforaphane 53-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 91-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 109-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 109-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 109-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-164 15090468-9 2004 Conversely, the induction of UGT1A1 transcription by sulforaphane was totally abolished by PD98059, although PD98059 slightly enhanced (20%) the induction of UGT1A1 by apigenin implying that the induction of UGT1A1 by sulforaphane is mediated by MAPK/extracellular signal-regulated kinase kinase, whereas UGT1A1 induction by apigenin may be associated with NF-kappaB translocation since the NF-kappaB translocation inhibitor, SN50 enhanced the induction of UGT by apigenin. sulforaphane 218-230 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 56-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 56-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 56-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 195-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-28 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 195-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 15090468-10 2004 The results show that UGT1A1 and GSTA1 are regulated by sulforaphane through different signal transduction pathways and the differences in the mechanisms of modulation of UGT1A1 transcription by sulforaphane and apigenin resulted in a synergistic effect between these two compounds in the induction of UGT1A1. sulforaphane 195-207 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-177 15560369-1 2004 Human UDP-glucuronosyltransferase (UGT) 1A1 is only enzyme in the conjugation of bilirubin for prevention of hyperbilirubinemia and jaundice. Bilirubin 81-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-43 15560369-10 2004 This region contains HNF1 element, therefore, we speculated that dexamethasone directly and/or indirectly stimulates UGT1A1 expression through this HNF1 region in the promoter region of UGT1A1. Dexamethasone 65-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 15560369-10 2004 This region contains HNF1 element, therefore, we speculated that dexamethasone directly and/or indirectly stimulates UGT1A1 expression through this HNF1 region in the promoter region of UGT1A1. Dexamethasone 65-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 15560369-11 2004 Thus, we clarified that UGT1A1 was induced by dexamethasone and the key position was the region (-97/-53) in UGT1A1 promoter. Dexamethasone 46-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 15560369-11 2004 Thus, we clarified that UGT1A1 was induced by dexamethasone and the key position was the region (-97/-53) in UGT1A1 promoter. Dexamethasone 46-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 15280927-1 2004 SN-38 is the active metabolite of irinotecan and it is metabolised through conjugation by uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 15280927-1 2004 SN-38 is the active metabolite of irinotecan and it is metabolised through conjugation by uridine diphosphate glucuronosyl transferase (UGT1A1). Irinotecan 34-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 15280927-2 2004 The major toxicity of irinotecan therapy is diarrhoea, which has been related to the enzymatic activity of UGT1A1. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 15280927-13 2004 The role of the UGT1A1 genotype as a predictor of toxicity in cancer patients receiving irinotecan demands the performance of a randomized trial to ascertain whether genotype-adjusted dosages of the drug can help to establish safe and effective doses not only for patients with the UGT1A1(*)28 homozygous genotype but also for those with the most common UGT1A1 6/6 or 6/7 genotype. Irinotecan 88-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 15357915-8 2004 The most potent inhibition was observed for the reaction between 17alpha-ethynylestradiol and UGT1A1 (K(i) = 10.5 microM), and the weakest interaction was detected for acetaminophen and UGT1A9 (IC(50) > 1 mM). Ethinyl Estradiol 65-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 15258113-4 2004 Of these UGTs, UGT1A1, 1A9, and 2B7, which are constitutively expressed in human livers, showed remarkable activity toward 8-OH-2,3,7-triCDD. 3,7,8-trichlorooxanthren-2-ol 123-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 15297419-0 2004 Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Irinotecan 47-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 306-316 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-95 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 306-316 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-104 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 318-324 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-95 15297419-1 2004 PURPOSE: We wanted to assess polymorphisms in the uridine diphosphoglucuronosyl transferase 1A1 (UGT 1A1) gene: the TATA box polymorphism and UGT 1A1 G71R and Y486D mutations in the coding sequence, the main mutations characterizing Gilbert"s syndrome, as predictors of severe toxic event occurrence after irinotecan (CPT-11) administration. Irinotecan 318-324 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-104 15310245-0 2004 Human UDP-glucuronosyltransferase 1A1 is the primary enzyme responsible for the N-glucuronidation of N-hydroxy-PhIP in vitro. Nitrogen 80-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-37 15310245-0 2004 Human UDP-glucuronosyltransferase 1A1 is the primary enzyme responsible for the N-glucuronidation of N-hydroxy-PhIP in vitro. 2-hydroxyamino-1-methyl-6-phenylimidazo(4,5-b)pyridine 101-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-37 15310245-1 2004 UDP-glucuronosyltransferase 1A proteins (UGT1A) catalyze the glucuronidation of many endogenous and xenobiotic compounds including heterocyclic amines and their hydroxylated metabolites. heterocyclic amines 131-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-30 15310245-7 2004 UGT1A1 was the most efficient in converting N-hydroxy-PhIP to both conjugates producing five times more of the N(2)-conjugate than UGT1A4, the next most active UGT, and 286 times more than UGT1A7, the least active UGT. 2-hydroxyamino-1-methyl-6-phenylimidazo(4,5-b)pyridine 44-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15310245-7 2004 UGT1A1 was the most efficient in converting N-hydroxy-PhIP to both conjugates producing five times more of the N(2)-conjugate than UGT1A4, the next most active UGT, and 286 times more than UGT1A7, the least active UGT. Nitrogen 111-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15310245-8 2004 With an apparent K(m) of 52 microM and a K(cat) of 114 min(-)(1), UGT1A1 was also the most catalytically efficient in forming N-hydroxy-PhIP-N(2)-glucuronide. n-hydroxy-phip-n(2)-glucuronide 126-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 15310245-9 2004 The catalytic efficiency for N-hydroxy-PhIP-N3-glucuronide formation was 8, 10, and 6 times lower for UGT1A1, -1A4, and -1A8, respectively, when compared to the K(cat) values for N-hydroxy-PhIP-N(2)-glucuronide formation. n-hydroxy-phip-n3-glucuronide 29-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 15310245-9 2004 The catalytic efficiency for N-hydroxy-PhIP-N3-glucuronide formation was 8, 10, and 6 times lower for UGT1A1, -1A4, and -1A8, respectively, when compared to the K(cat) values for N-hydroxy-PhIP-N(2)-glucuronide formation. n-hydroxy-phip-n(2)-glucuronide 179-210 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 15310245-10 2004 These results clearly show that UGT1A1 has the highest specificity for glucuronidating N-hydroxy-PhIP. 2-hydroxyamino-1-methyl-6-phenylimidazo(4,5-b)pyridine 87-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 15286088-0 2004 Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. Irinotecan 85-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15122071-5 2004 Only minor glucuronidation was observed for thiocoraline by UGT1A1 and UGT1A9 and no glucuronidation was observed in human liver S9 fraction. thiocoraline 44-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 15286088-1 2004 The uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A9 isoforms are involved in the phase II biotransformation of the irinotecan metabolite, SN-38. Irinotecan 129-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-65 15286088-8 2004 The median AUC ratio of SN-38G to SN-38 was significantly reduced in carriers of the variant UGT1A1(*)28 allele (7.00 [Wt] vs. 6.26 [Het] vs. 2.51 [Var]; p =.022). Irinotecan 24-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 15286088-1 2004 The uridine diphosphate glucuronosyltransferase (UGT) 1A1 and 1A9 isoforms are involved in the phase II biotransformation of the irinotecan metabolite, SN-38. Irinotecan 152-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-65 15286088-8 2004 The median AUC ratio of SN-38G to SN-38 was significantly reduced in carriers of the variant UGT1A1(*)28 allele (7.00 [Wt] vs. 6.26 [Het] vs. 2.51 [Var]; p =.022). Irinotecan 34-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 15286088-10 2004 Screening for the UGT1A1(*)28 polymorphism may identify patients with altered SN-38 pharmacokinetics. Irinotecan 78-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 15286088-3 2004 The aim of this study was to evaluate the functional consequence of the UGT1A1(TA)(7)TAA (UGT1A1(*)28), UGT1A9 766G>A (D256N; UGT1A9(*)5), and UGT1A9 98T>C (M33T; UGT1A9(*)3) variants in Caucasian patients treated with irinotecan. Irinotecan 225-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 15099818-0 2004 Bilirubin UDP-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphisms and HPRT, glycophorin A, and micronuclei mutant frequencies in human blood. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-41 15179405-0 2004 UGT1A1 haplotypes associated with reduced glucuronidation and increased serum bilirubin in irinotecan-administered Japanese patients with cancer. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15179405-0 2004 UGT1A1 haplotypes associated with reduced glucuronidation and increased serum bilirubin in irinotecan-administered Japanese patients with cancer. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 15179405-10 2004 CONCLUSION: This study identified several UGT1A1 haplotypes significantly associated with the reduced AUC ratio (*28 and *6) and with the increased total bilirubin level (*28, *60, and *IB) and suggested that the novel haplotype *IB might be functionally important. Bilirubin 154-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 15254716-1 2004 (Uridino-diphosphate)glucuronosyl-transferase enzyme 1A1 isoform (UGT1A1) is involved in glucuronidation of antineoplastic drugs such as SN38, the active metabolite of irinotecan, as well as estrogens and their metabolites. Irinotecan 168-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 15135306-7 2004 Only UGT1A4 catalyzed the N-linked glucuronidation of TAM among recombinant UGTs (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B4, UGT2B7, UGT2B15, and UGT2B17) expressed in insect cells. Tamoxifen 54-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 15155549-10 2004 Silybin was a potent inhibitor of UGT1A1 and was 14- and 20-fold more selective for UGT1A1 than for UGT1A9 and UGT1A6, respectively. Silybin 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 15155549-10 2004 Silybin was a potent inhibitor of UGT1A1 and was 14- and 20-fold more selective for UGT1A1 than for UGT1A9 and UGT1A6, respectively. Silybin 0-7 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 15099818-0 2004 Bilirubin UDP-glucuronosyltransferase 1A1 (UGT1A1) gene promoter polymorphisms and HPRT, glycophorin A, and micronuclei mutant frequencies in human blood. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 15099818-1 2004 A dinucleotide repeat polymorphism (5-, 6-, 7-, or 8-TA units) has been identified within the promoter region of UDP-glucuronosyltransferase 1A1 (UGT1A1) gene. Dinucleoside Phosphates 2-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-144 15099818-1 2004 A dinucleotide repeat polymorphism (5-, 6-, 7-, or 8-TA units) has been identified within the promoter region of UDP-glucuronosyltransferase 1A1 (UGT1A1) gene. Dinucleoside Phosphates 2-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 15099818-4 2004 Because bilirubin is a known antioxidant, we hypothesized that UGT1A1 repeats associated with higher bilirubin may be protective against oxidative damage. Bilirubin 8-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 15099818-4 2004 Because bilirubin is a known antioxidant, we hypothesized that UGT1A1 repeats associated with higher bilirubin may be protective against oxidative damage. Bilirubin 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 15007088-0 2004 Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. Irinotecan 103-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-55 15007088-2 2004 UDP-glucuronosyltransferase 1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38. Irinotecan 96-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 15007088-2 2004 UDP-glucuronosyltransferase 1A1 (UGT1A1) catalyzes the glucuronidation of the active metabolite SN-38. Irinotecan 96-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 15007088-14 2004 CONCLUSION: UGT1A1 genotype and total bilirubin levels are strongly associated with severe neutropenia, and could be used to identify cancer patients predisposed to the severe toxicity of irinotecan. Irinotecan 188-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 15104253-0 2004 The induction of human UDP-glucuronosyltransferase 1A1 mediated through a distal enhancer module by flavonoids and xenobiotics. Flavonoids 100-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-54 15104253-1 2004 We identified the UDP-glucuronosyltransferase (UGT) 1A1 5"-upstream region that confers UGT1A1 induction by various agents, including flavonoids, on a luciferase reporter gene and has the properties of a transcriptional enhancer. Flavonoids 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-55 15104253-1 2004 We identified the UDP-glucuronosyltransferase (UGT) 1A1 5"-upstream region that confers UGT1A1 induction by various agents, including flavonoids, on a luciferase reporter gene and has the properties of a transcriptional enhancer. Flavonoids 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15104253-3 2004 Utilizing transactivation experiments with the UGT1A1 290-bp reporter gene, we assessed UGT1A1 induction by various flavonoids. Flavonoids 116-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 15104253-5 2004 The treatment of HepG2 cells with the flavonoids for 24 hr elevated the expression of mRNAs and proteins of UGT1A1 and CYP1A1, while the mRNA levels of CYP2B6, CYP3A4, CAR, PXR and AhR was not altered. Flavonoids 38-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 15104253-8 2004 Taken together, the results indicate that UGT1A1 was induced in response to flavonoids and xenobiotics through the transactivation of the 290-bp reporter gene, that was a multi-component enhancer containing CAR, PXR and AhR motifs. Flavonoids 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 14977865-9 2004 In conclusion, the formation of SCH 60663 is mediated via UGT1A1, UGT1A3, and UGT2B15, and the formation SCH 488128 is mediated via UGT2B7. SCH 60663 32-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 12969965-1 2004 Etoposide is a substrate for P-glycoprotein, CYP3A4, CYP3A5, and UGT1A1. Etoposide 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 14977865-7 2004 Recombinant human UGT1A1, UGT1A3, and UGT2B15 all exhibited catalytic activity with respect to the formation of the phenolic glucuronide. Glucuronides 125-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 14744946-0 2004 Involvement of human hepatic UGT1A1, UGT2B4, and UGT2B7 in the glucuronidation of carvedilol. Carvedilol 82-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 14744946-4 2004 The glucuronidation of carvedilol was catalyzed by at least three recombinant UGT isoforms: UGT1A1, UGT2B4, and UGT2B7. Carvedilol 23-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 14744946-5 2004 UGT2B4 formed both G1 and G2, whereas UGT1A1 and UGT2B7 were responsible for the formation of glucuronide G2 and G1, respectively. Glucuronides 94-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 14744946-6 2004 The enzyme kinetics for carvedilol glucuronidation by UGT1A1, UGT2B4, and UGT2B7 in addition to human liver microsomes were examined by Lineweaver-Burk analysis. Carvedilol 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 14978227-1 2004 Human constitutive androstane (or active) receptor (hCAR), a member of the nuclear receptor superfamily NR1I3, regulates the expression of several genes that are mainly involved in the metabolism of endogenous and xenobiotic compounds (e.g., CYP2B6, CYP3A4, and UGT1A1). androstane 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 262-268 14978227-7 2004 In the reporter gene assays employing the phenobarbital-responsible enhancer module (PBREM) from CYP2B6 and UGT1A1 genes, the splice variants, except for SV1, were inactive, whereas SV1 transactivated the CYP2B6 PBREM but not the UGT1A1 PBREM reporter. Phenobarbital 42-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 14978227-7 2004 In the reporter gene assays employing the phenobarbital-responsible enhancer module (PBREM) from CYP2B6 and UGT1A1 genes, the splice variants, except for SV1, were inactive, whereas SV1 transactivated the CYP2B6 PBREM but not the UGT1A1 PBREM reporter. Phenobarbital 42-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 230-236 14871858-1 2004 UDP-glucuronosyltransferase (UGT) 1A1 is involved in the inactivation of estradiol (E(2)) and its oxidized metabolites. Estradiol 73-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 14871858-5 2004 Intrinsic clearances indicated that UGT1A1 had a preference for the glucuronidation of 2-hydroxyestradiol, a metabolite associated with antiproliferative activity. 2-hydroxyestradiol 87-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 36-42 14871858-11 2004 These observations suggest that lower expression of UGT1A1 decreases the risk of endometrial cancer by reducing the excretion of 2-hydroxyestradiol, the antiproliferative metabolite of E(2), in the endometrium. 2-hydroxyestradiol 129-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 14871858-11 2004 These observations suggest that lower expression of UGT1A1 decreases the risk of endometrial cancer by reducing the excretion of 2-hydroxyestradiol, the antiproliferative metabolite of E(2), in the endometrium. Estradiol 185-189 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 15180166-2 2004 The underlying cause of GS is a polymorphism in the promotor region of the uridine diphosphate glucuronosyltransferase isoform 1A1 gene (UGT1A1*28), its encoded enzyme being responsible for the glucuronidation of bilirubin and presumably acetaminophen. Bilirubin 213-222 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 15180166-2 2004 The underlying cause of GS is a polymorphism in the promotor region of the uridine diphosphate glucuronosyltransferase isoform 1A1 gene (UGT1A1*28), its encoded enzyme being responsible for the glucuronidation of bilirubin and presumably acetaminophen. Acetaminophen 238-251 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 15180166-4 2004 Patients with GS might be more susceptible to unexpected side effects while taking acetaminophen and other drugs which are substrates of UGT1A1. Acetaminophen 83-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 15180166-9 2004 The metabolic ratios unchanged acetaminophen/acetaminophen glucuronide in UGT1A1-wildtypes, heterozygotes and mutants showed no statistically significant differences. Acetaminophen 31-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 12969965-10 2004 The UGT1A1 6/6 genotype predicted lower catechol AUC. catechol 40-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 14744740-0 2004 Correlation between the UDP-glucuronosyltransferase (UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype: significantly decreased glucuronidating activity against benzo(a)pyrene-7,8-dihydrodiol(-) in liver microsomes from subjects with the UGT1A1*28 variant. benzo 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 14744740-0 2004 Correlation between the UDP-glucuronosyltransferase (UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype: significantly decreased glucuronidating activity against benzo(a)pyrene-7,8-dihydrodiol(-) in liver microsomes from subjects with the UGT1A1*28 variant. benzo 182-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 259-265 14744740-0 2004 Correlation between the UDP-glucuronosyltransferase (UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype: significantly decreased glucuronidating activity against benzo(a)pyrene-7,8-dihydrodiol(-) in liver microsomes from subjects with the UGT1A1*28 variant. pyrene-7,8-dihydrodiol 190-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 53-59 14744740-1 2004 Of the hepatic UDP-glucuronosyltransferases (UGTs), only UGT1A1 and UGT1A9 exhibit activity against benzo(a)pyrene-trans-7R,8R-dihydrodiol [BPD(-)], precursor to the highly mutagenic anti-(+)-benzo(a)pyrene-7R,8S-dihydrodiol-9S,10R-epoxide. -trans-7r,8r-dihydrodiol 114-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 14744740-1 2004 Of the hepatic UDP-glucuronosyltransferases (UGTs), only UGT1A1 and UGT1A9 exhibit activity against benzo(a)pyrene-trans-7R,8R-dihydrodiol [BPD(-)], precursor to the highly mutagenic anti-(+)-benzo(a)pyrene-7R,8S-dihydrodiol-9S,10R-epoxide. benzo(a)pyrene 7,8-dihydrodiol 140-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 14744740-1 2004 Of the hepatic UDP-glucuronosyltransferases (UGTs), only UGT1A1 and UGT1A9 exhibit activity against benzo(a)pyrene-trans-7R,8R-dihydrodiol [BPD(-)], precursor to the highly mutagenic anti-(+)-benzo(a)pyrene-7R,8S-dihydrodiol-9S,10R-epoxide. (+)-benzo 188-197 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 14744740-1 2004 Of the hepatic UDP-glucuronosyltransferases (UGTs), only UGT1A1 and UGT1A9 exhibit activity against benzo(a)pyrene-trans-7R,8R-dihydrodiol [BPD(-)], precursor to the highly mutagenic anti-(+)-benzo(a)pyrene-7R,8S-dihydrodiol-9S,10R-epoxide. pyrene-7r,8s-dihydrodiol-9s,10r-epoxide 200-239 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Dinucleoside Phosphates 58-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Dinucleoside Phosphates 58-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Dinucleoside Phosphates 58-70 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Bilirubin 219-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Bilirubin 219-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 14744740-2 2004 The UGT1A1*28 allelic variant contains an additional (TA) dinucleotide repeat in the "TATAA" box [(TA)(6)>(TA)(7)] of the UGT1A1 promoter that has been linked to decreased expression of the UGT1A1 gene and decreased bilirubin conjugation, leading to the relatively nondebilitating condition known as Gilbert"s syndrome. Bilirubin 219-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 14744740-4 2004 Significant decreases in UGT1A1 protein (P < 0.005) and bilirubin conjugation activity (P < 0.001) were observed in liver microsomes from subjects homozygous for the UGT1A1*28 allelic variant compared with subjects homozygous for the wild-type UGT1A1*1 allele. Bilirubin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-178 14744740-4 2004 Significant decreases in UGT1A1 protein (P < 0.005) and bilirubin conjugation activity (P < 0.001) were observed in liver microsomes from subjects homozygous for the UGT1A1*28 allelic variant compared with subjects homozygous for the wild-type UGT1A1*1 allele. Bilirubin 59-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-178 14744740-5 2004 Significant decreases in BPD(-) glucuronidation activity (P < 0.02) were observed in subjects with the UGT1A1(*28/*28) genotype compared with subjects having the wild-type UGT1A1(*1/*1) genotype in assays of liver microsomes that included 0.1 mM alpha-naphthylamine, a competitive inhibitor of UGT1A9 and not UGT1A1. 1-Naphthylamine 249-268 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 14744740-8 2004 These data suggest that the UGT1A1 TATAA box polymorphism plays a role in an individual"s overall ability to detoxify benzo(a)pyrene and in cancer risk. benzo(a) 118-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 14744740-8 2004 These data suggest that the UGT1A1 TATAA box polymorphism plays a role in an individual"s overall ability to detoxify benzo(a)pyrene and in cancer risk. pyrene 126-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 14633881-0 2003 Haplotype structure of the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene and its relationship to serum total bilirubin concentration in a male Korean population. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-58 12949046-6 2003 In contrast, SFN induced phase II detoxification enzymes, UDP-glucuronosyltransferase 1A1 and glutathione S-transferase A1 mRNA expression. sulforaphane 13-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-89 14633881-0 2003 Haplotype structure of the UDP-glucuronosyltransferase 1A1 (UGT1A1) gene and its relationship to serum total bilirubin concentration in a male Korean population. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 12914384-8 2003 UGT1A1 (uridine diphoshate-glucuronosyltransferase 1A1) is involved on irinotecan metabolism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 14586211-0 2003 Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 118-165 14586211-0 2003 Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Irinotecan 18-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 14586211-0 2003 Weekly regimen of irinotecan/docetaxel in previously treated non-small cell lung cancer patients and correlation with uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism. Docetaxel 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 14550264-1 2003 UDP-glucuronosyltransferase form 1A1 (UGT1A1) is the only bilirubin-glucuronidating isoform of this protein, and genetic deficiencies of UGT1A1 cause Crigler-Najjar syndrome, a disorder resulting from nonhemolytic unconjugated hyperbilirubinemia. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-36 14550264-1 2003 UDP-glucuronosyltransferase form 1A1 (UGT1A1) is the only bilirubin-glucuronidating isoform of this protein, and genetic deficiencies of UGT1A1 cause Crigler-Najjar syndrome, a disorder resulting from nonhemolytic unconjugated hyperbilirubinemia. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 14550264-1 2003 UDP-glucuronosyltransferase form 1A1 (UGT1A1) is the only bilirubin-glucuronidating isoform of this protein, and genetic deficiencies of UGT1A1 cause Crigler-Najjar syndrome, a disorder resulting from nonhemolytic unconjugated hyperbilirubinemia. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 14550264-2 2003 Here we have focused on the instability of a translocation-deficient UGT1A1 protein, which has been found in patients with Crigler-Najjar type II, to elucidate the molecular basis underlying the deficiency in glucuronidation of bilirubin. Bilirubin 228-237 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 69-75 14555337-6 2003 Studies aimed at identifying the human liver UDP-glucuronosyltransferase (UGT) enzyme(s) involved in the glucuronidation of bropirimine were carried out using recombinant human UGTs and it was determined that glucuronidation of bropirimine was catalysed by UGT1A1, UGT1A3 and UGT1A9. bropirimine 124-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 257-263 14555337-12 2003 The results demonstrate that UGT1A9 and to a lesser extent UGT1A1 are responsible for the majority of bropirimine O-glucuronidation in man. bropirimine 102-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 14705859-2 2003 The orphan nuclear receptor CAR (NR1I3 controls phase I (CYP2B, CYP2C, CYP3A), phase II (UGT1A1), and transporter (SLC21A6, MRP2) genes involved in drug metabolism and bilirubin clearance. Bilirubin 168-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 12914384-8 2003 UGT1A1 (uridine diphoshate-glucuronosyltransferase 1A1) is involved on irinotecan metabolism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-54 12730278-3 2003 Although several genetic polymorphisms are reported for UGT1A1 and UGT1A7 that affect the SN-38 glucuronidation activities, no such polymorphisms have been identified for UGT1A9. Irinotecan 90-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 12960109-7 2003 The extent of SN-38 glucuronidation was slightly impaired in homozygous variants of UGT1A1*28, although differences were not statistically significant (P = 0.22). Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 12867491-0 2003 Glucuronidation of HMR1098 in human microsomes: evidence for the involvement of UGT1A1 in the formation of S-glucuronides. Glucuronides 107-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 12850481-2 2003 The molecular basis of this syndrome usually concerns an additional dinucleotide insertion (TA) in the A(TA)(n)TAA configuration residing in the promoter region of the UGT1 A1 gene. Dinucleoside Phosphates 68-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 168-175 12850492-1 2003 The promoter region of the UDP glucuronosyltransferase 1 gene (UGT1A1) contains a run of thymine-adenine (TA) repeats, usually six (TA)(6). thymine-adenine 89-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12850492-1 2003 The promoter region of the UDP glucuronosyltransferase 1 gene (UGT1A1) contains a run of thymine-adenine (TA) repeats, usually six (TA)(6). Tantalum 106-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12756216-8 2003 The changes in gene expression were also seen in differentiated Caco-2 cells, where sulforaphane was responsible for induction of GSTA1 and quercetin for induction of UGT1A1. sulforaphane 84-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 12756209-8 2003 Nontraditional kinetics were also observed when other UGT1A1 substrates such as ethinylestradiol, buprenorphine, and anthraflavic acid were studied with both human liver microsomes and recombinant UGT1A1. 2,6-dihydroxyanthraquinone 117-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 197-203 12756216-8 2003 The changes in gene expression were also seen in differentiated Caco-2 cells, where sulforaphane was responsible for induction of GSTA1 and quercetin for induction of UGT1A1. Quercetin 140-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 12695347-0 2003 Glucuronidation of etoposide in human liver microsomes is specifically catalyzed by UDP-glucuronosyltransferase 1A1. Etoposide 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-115 12695347-6 2003 The kinetic parameters of etoposide glucuronidation were K(m) = 439.6 +/- 70.7 microM and V(max) = 255.6 +/- 19.2 pmol/min/mg of protein in human liver microsomes and K(m) = 503.2 +/- 110.2 microM and V(max) = was 266.5 +/- 28.6 pmol/min/mg of protein in recombinant UGT1A1. Etoposide 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 267-273 12695347-7 2003 The etoposide glucuronidation in pooled human liver microsomes was inhibited by bilirubin (IC(50) = 31.7 microM) and estradiol (IC(50) = 34 microM) as typical substrates for UGT1A1. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 174-180 12769780-8 2003 In particular, the UGT1A1*28 allele has been associated with an increased toxicity after irinotecan chemotherapy. Irinotecan 89-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 12695347-11 2003 These results demonstrate that etoposide glucuronidation in human liver microsomes is specifically catalyzed by UGT1A1. Etoposide 31-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 112-118 12800608-0 2003 Irinotecan treatment in cancer patients with UGT1A1 polymorphisms. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 54-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-6 2003 Two recent pharmacogenetic trials (one performed in the United States and the other in Japan) investigated the clinical significance of UGT1A1 gene mutations for both the pharmacokinetics of irinotecan metabolites and the toxicity profile. Irinotecan 191-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 66-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-1 2003 At the present time, pharmacogenetic investigation of irinotecan (CPT-11, Camptosar) therapy is mainly focused on the clinical relevance of genetic variation in the UDP-glucuronosyltransferase (UGT1A1) gene. Irinotecan 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 194-200 12800608-3 2003 UGT1A1 genotypes associated with Gilbert"s syndrome (a mild intermittent hyperbilirubinemia) are characterized by reduced glucuronidation of SN-38. Irinotecan 141-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12480553-4 2003 UGT1A1*28 is associated with Gilbert"s syndrome, a deficiency in glucuronidation of bilirubin leading to mild hyperbilirubinemia, whereas UGT1A6*2 may result in low glucuronidation rates of several drugs. Bilirubin 84-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12435745-3 2003 The activity of recombinant UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT2B4, UGT2B7, and UGT2B15 was almost fully inhibited by 0.2% Triton X-100. Octoxynol 123-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 12566446-1 2003 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an important physiological role by contributing to the metabolism of endogenous substances such as bilirubin in addition to xenobiotics and drugs. Bilirubin 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 12566446-1 2003 UDP-glucuronosyltransferase 1A1 (UGT1A1) plays an important physiological role by contributing to the metabolism of endogenous substances such as bilirubin in addition to xenobiotics and drugs. Bilirubin 146-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 12566446-3 2003 In this report, we show that in human hepatoma HepG2 cells the UGT1A1 gene is also inducible with aryl hydrocarbon receptor (Ah receptor) ligands such as 2,3,7,8-tetrachlodibenzo-p-dioxin (TCDD), beta-naphthoflavone, and benzo[a]pyrene metabolites. 2,3,7,8-tetrachlodibenzo-p-dioxin 154-187 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12566446-3 2003 In this report, we show that in human hepatoma HepG2 cells the UGT1A1 gene is also inducible with aryl hydrocarbon receptor (Ah receptor) ligands such as 2,3,7,8-tetrachlodibenzo-p-dioxin (TCDD), beta-naphthoflavone, and benzo[a]pyrene metabolites. Polychlorinated Dibenzodioxins 189-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12566446-3 2003 In this report, we show that in human hepatoma HepG2 cells the UGT1A1 gene is also inducible with aryl hydrocarbon receptor (Ah receptor) ligands such as 2,3,7,8-tetrachlodibenzo-p-dioxin (TCDD), beta-naphthoflavone, and benzo[a]pyrene metabolites. beta-Naphthoflavone 196-215 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12566446-3 2003 In this report, we show that in human hepatoma HepG2 cells the UGT1A1 gene is also inducible with aryl hydrocarbon receptor (Ah receptor) ligands such as 2,3,7,8-tetrachlodibenzo-p-dioxin (TCDD), beta-naphthoflavone, and benzo[a]pyrene metabolites. Benzo(a)pyrene 221-235 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12566446-8 2003 Gel mobility shift analysis confirmed that TCDD induction of nuclear proteins specifically bound to the UGT1A1-XRE, and competition experiments with Ah receptor and Arnt antibodies demonstrated that the nuclear protein was the Ah receptor. Polychlorinated Dibenzodioxins 43-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 12618960-7 2003 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid and thus enhances bilirubin elimination; therefore, it is an important candidate gene for serum bilirubin. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12618960-7 2003 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid and thus enhances bilirubin elimination; therefore, it is an important candidate gene for serum bilirubin. Glucuronic Acid 51-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12618960-7 2003 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid and thus enhances bilirubin elimination; therefore, it is an important candidate gene for serum bilirubin. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12618960-7 2003 UGT1A1 catalyzes the conjugation of bilirubin with glucuronic acid and thus enhances bilirubin elimination; therefore, it is an important candidate gene for serum bilirubin. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12618960-10 2003 Our findings suggest that UGT1A1 may be a major gene controlling serum bilirubin levels in the population. Bilirubin 71-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 12642472-8 2003 Human UGT1A1, 1A8, and 1A9 had high activities with EGCG. epigallocatechin gallate 52-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 12644700-1 2003 Through a multiplex promoter spanning 218 kb, the phase II UDP-glucuronosyltransferase 1A (UGT1) gene encodes at least eight differently regulated mRNAs whose protein products function as the principal means to eliminate a vast array of steroids, heme metabolites, environmental toxins, and drugs. Steroids 237-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-89 12644700-1 2003 Through a multiplex promoter spanning 218 kb, the phase II UDP-glucuronosyltransferase 1A (UGT1) gene encodes at least eight differently regulated mRNAs whose protein products function as the principal means to eliminate a vast array of steroids, heme metabolites, environmental toxins, and drugs. Steroids 237-245 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-95 12644700-1 2003 Through a multiplex promoter spanning 218 kb, the phase II UDP-glucuronosyltransferase 1A (UGT1) gene encodes at least eight differently regulated mRNAs whose protein products function as the principal means to eliminate a vast array of steroids, heme metabolites, environmental toxins, and drugs. Heme 247-251 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-89 12644700-1 2003 Through a multiplex promoter spanning 218 kb, the phase II UDP-glucuronosyltransferase 1A (UGT1) gene encodes at least eight differently regulated mRNAs whose protein products function as the principal means to eliminate a vast array of steroids, heme metabolites, environmental toxins, and drugs. Heme 247-251 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-95 12646172-2 2003 Likewise, bilirubin-UGT1A1 expressed in COS-1 cells was inhibited by curcumin and calphostin-C. Bilirubin 10-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 12646172-2 2003 Likewise, bilirubin-UGT1A1 expressed in COS-1 cells was inhibited by curcumin and calphostin-C. Curcumin 69-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 12646172-2 2003 Likewise, bilirubin-UGT1A1 expressed in COS-1 cells was inhibited by curcumin and calphostin-C. calphostin C 82-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 12646172-4 2003 UGT1A1 incorporated [33P]orthophosphate, which was inhibited by calphostin-C. Phosphate-33P (9CI) 20-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12646172-4 2003 UGT1A1 incorporated [33P]orthophosphate, which was inhibited by calphostin-C. Carbon 75-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 12570720-6 2003 Irinotecan is subjected to be shunted between CYP3A4 mediated oxidative metabolism to form two inactive metabolites APC or NPC and tissue carboxylesterase mediated hydrolysis to form SN-38 which is eventually detoxified via glucuronidation by UGT1A1 to form SN-38G. Irinotecan 183-188 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 243-249 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 19-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 83-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-0 2003 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of irinotecan (CPT-11), by human UGT1A1 variants, G71R, P229Q, and Y486D. Irinotecan 95-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 0-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 32-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. Irinotecan 92-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-1 2003 7-Ethyl-10-hydroxycamptothecin (SN-38), an active metabolite of antitumor agent irinotecan (CPT-11), is conjugated and detoxified to SN-38-glucuronide by UDP-glucuronosyltransferase (UGT) 1A1. 7-ethyl-10-hydroxycamptothecin glucuronide 133-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 154-191 12485959-2 2003 Genetic polymorphisms in UGT1A1 are thought to contribute to severe diarrhea and/or leukopenia caused by CPT-11. Irinotecan 105-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 12485959-4 2003 However, little information is available on the influence of UGT1A1 polymorphisms in the coding region on the SN-38 glucuronidation activity. Irinotecan 110-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-67 12485959-5 2003 In the present study, wild-type (WT) and three variant (G71R, P229Q, and Y486D) cDNAs of human UGT1A1s were transiently expressed in COS-1 cells, and the kinetic parameters of these UGT1A1s were determined for SN-38 glucuronidation. carbonyl sulfide 133-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 12485959-6 2003 A partially reduced UGT1A1 protein expression was observed in COS-1 cells for G71R and Y486D. carbonyl sulfide 62-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 12485959-7 2003 WT UGT1A1 catalyzed SN-38 glucuronidation with an apparent K(m) value of 11.5 microM, whereas those of G71R, P229Q, and Y486D were 14.0, 18.0, and 63.5 microM, respectively. Irinotecan 20-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 3-9 12485959-10 2003 The decreased SN-38 glucuronidation efficiency ratio of G71R and P229Q could be critical in combination with other polymorphisms in the UGT1A1 gene. Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 12480553-9 2003 CONCLUSIONS: Most patients with Gilbert"s syndrome, in addition to their reduced B-UGT enzyme activity, may have abnormalities in the glucuronidation of aspirin or coumarin- and dopamine-derivatives, due to this combination of UGT1A1*28 and UGT1A6*2 genotypes. Aspirin 153-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 227-233 12433820-4 2002 The kinetics of the troglitazone glucuronidation in the recombinant UGT1A10 and UGT1A1 exhibited an atypical pattern of substrate inhibition when the substrate concentration was over 200 micro M. With a Michaelis-Menten equation at 6 to 200 micro M troglitazone, the K(m) value was 11.1 +/- 5.8 micro M and the V(max) value was 33.6 +/- 3.7 pmol/min/mg protein in recombinant UGT1A10. Troglitazone 20-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 12466075-7 2002 Patients with the UGT1A1 TA*7/TA*7 genotype showed higher mean serum bilirubin levels than did patients who were homozygous for the wild-type allele (Mann-Whitney test 35.5; p < 0.05). Bilirubin 69-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 12433804-2 2002 Although UGT1A1 uniquely catalyzes the glucuronidation of the endobiotic, bilirubin, and UGT2B7 uniquely catalyzes the glucuronidation of morphine to both the 3-0 glucuronide and the 6-0 glucuronide, both catalyze the glucuronidation of the mixed opioid agonist/antagonist buprenorphine with high efficiency. Bilirubin 74-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 12433804-2 2002 Although UGT1A1 uniquely catalyzes the glucuronidation of the endobiotic, bilirubin, and UGT2B7 uniquely catalyzes the glucuronidation of morphine to both the 3-0 glucuronide and the 6-0 glucuronide, both catalyze the glucuronidation of the mixed opioid agonist/antagonist buprenorphine with high efficiency. Morphine 138-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 12433823-10 2002 In conclusion, the involvement of UGT1A1 and UGT1A9 as well as UGT1A4 in nicotine and cotinine N-glucuronidations in human liver microsomes was suggested, although the contributions of each UGT isoform could not be determined conclusively. Cotinine 86-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 12433820-4 2002 The kinetics of the troglitazone glucuronidation in the recombinant UGT1A10 and UGT1A1 exhibited an atypical pattern of substrate inhibition when the substrate concentration was over 200 micro M. With a Michaelis-Menten equation at 6 to 200 micro M troglitazone, the K(m) value was 11.1 +/- 5.8 micro M and the V(max) value was 33.6 +/- 3.7 pmol/min/mg protein in recombinant UGT1A10. Troglitazone 249-261 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 68-74 12433823-10 2002 In conclusion, the involvement of UGT1A1 and UGT1A9 as well as UGT1A4 in nicotine and cotinine N-glucuronidations in human liver microsomes was suggested, although the contributions of each UGT isoform could not be determined conclusively. Nitrogen 95-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 12433820-11 2002 The troglitazone glucuronosyltransferase activity in pooled human liver microsomes was strongly inhibited by bilirubin (IC(50) = 1.9 micro M), a typical substrate of UGT1A1. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 12433804-2 2002 Although UGT1A1 uniquely catalyzes the glucuronidation of the endobiotic, bilirubin, and UGT2B7 uniquely catalyzes the glucuronidation of morphine to both the 3-0 glucuronide and the 6-0 glucuronide, both catalyze the glucuronidation of the mixed opioid agonist/antagonist buprenorphine with high efficiency. 3-0 glucuronide 159-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 12433804-2 2002 Although UGT1A1 uniquely catalyzes the glucuronidation of the endobiotic, bilirubin, and UGT2B7 uniquely catalyzes the glucuronidation of morphine to both the 3-0 glucuronide and the 6-0 glucuronide, both catalyze the glucuronidation of the mixed opioid agonist/antagonist buprenorphine with high efficiency. 6-0 glucuronide 183-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 12433820-12 2002 These results suggested that the troglitazone glucuronidation in human liver would be mainly catalyzed by UGT1A1. Troglitazone 33-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 12433804-2 2002 Although UGT1A1 uniquely catalyzes the glucuronidation of the endobiotic, bilirubin, and UGT2B7 uniquely catalyzes the glucuronidation of morphine to both the 3-0 glucuronide and the 6-0 glucuronide, both catalyze the glucuronidation of the mixed opioid agonist/antagonist buprenorphine with high efficiency. Buprenorphine 273-286 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 12433823-7 2002 Nicotine and cotinine N-glucuronidations in pooled human liver microsomes were competitively inhibited by bilirubin as a substrate for UGT1A1 (K(i) = 3.9 and 3.3 micro M), imipramine as a substrate for UGT1A4 (K(i) = 6.1 and 2.7 micro M), and propofol as a substrate for UGT1A9 (K(i) = 6.0 and 12.0 micro M). Nicotine 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 12433804-5 2002 Etonitazenyl also competitively inhibited the glucuronidation of buprenorphine catalyzed by UGT1A1. etonitazenyl 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 12433804-5 2002 Etonitazenyl also competitively inhibited the glucuronidation of buprenorphine catalyzed by UGT1A1. Buprenorphine 65-78 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 12433823-7 2002 Nicotine and cotinine N-glucuronidations in pooled human liver microsomes were competitively inhibited by bilirubin as a substrate for UGT1A1 (K(i) = 3.9 and 3.3 micro M), imipramine as a substrate for UGT1A4 (K(i) = 6.1 and 2.7 micro M), and propofol as a substrate for UGT1A9 (K(i) = 6.0 and 12.0 micro M). cotinine n 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 12433804-8 2002 Anthraflavic acid and catechol estrogen glucuronidation, catalyzed by UGT1A1, was also not inhibited by etonitazenyl or buprenorphine. 2,6-dihydroxyanthraquinone 0-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 12433804-8 2002 Anthraflavic acid and catechol estrogen glucuronidation, catalyzed by UGT1A1, was also not inhibited by etonitazenyl or buprenorphine. catechol 22-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-76 12433823-7 2002 Nicotine and cotinine N-glucuronidations in pooled human liver microsomes were competitively inhibited by bilirubin as a substrate for UGT1A1 (K(i) = 3.9 and 3.3 micro M), imipramine as a substrate for UGT1A4 (K(i) = 6.1 and 2.7 micro M), and propofol as a substrate for UGT1A9 (K(i) = 6.0 and 12.0 micro M). Bilirubin 106-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 12433823-7 2002 Nicotine and cotinine N-glucuronidations in pooled human liver microsomes were competitively inhibited by bilirubin as a substrate for UGT1A1 (K(i) = 3.9 and 3.3 micro M), imipramine as a substrate for UGT1A4 (K(i) = 6.1 and 2.7 micro M), and propofol as a substrate for UGT1A9 (K(i) = 6.0 and 12.0 micro M). Propofol 243-251 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 12433823-10 2002 In conclusion, the involvement of UGT1A1 and UGT1A9 as well as UGT1A4 in nicotine and cotinine N-glucuronidations in human liver microsomes was suggested, although the contributions of each UGT isoform could not be determined conclusively. Nicotine 73-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 12464801-2 2002 UGT1A1 basal transcription is affected by a polymorphic (TA)n repeat, and another important regulatory element is the phenobarbital-responsive enhancer module (PBREM) which might contain variants affecting inducible gene expression. Phenobarbital 118-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 12464801-4 2002 We also investigated the relationship between PBREM-(TA)n haplotypes and the glucuronidation rate of the UGT1A1 substrate SN-38. Irinotecan 122-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 105-111 12386134-0 2002 Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Estradiol 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-58 12386134-0 2002 Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Estradiol 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 12386132-0 2002 Involvement of multiple UDP-glucuronosyltransferase 1A isoforms in glucuronidation of 5-(4"-hydroxyphenyl)-5-phenylhydantoin in human liver microsomes. hydroxyphenytoin 86-124 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-54 12386134-0 2002 Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Estradiol 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 125-131 12386132-7 2002 The 4"-HPPH O-glucuronidation in pooled human liver microsomes was inhibited by beta-estradiol as a typical substrate for UGT1A1 (IC(50) = 21.1 microM) and imipramine as a typical substrate for UGT1A4 (IC(50) = 57.7 microM). Estradiol 80-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 12386134-2 2002 In confirmation of previous work using alamethicin-treated HLM pooled from four livers, UGT1A1-catalyzed estradiol-3-glucuronidation demonstrated homotropic activation kinetics (S(50) = 22 microM, Hill coefficient, n = 1.9) whereas estradiol-17-glucuronidation (catalyzed by other UGT enzymes) followed Michaelis-Menten kinetics (K(m) = 7 microM). Alamethicin 39-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 12386134-2 2002 In confirmation of previous work using alamethicin-treated HLM pooled from four livers, UGT1A1-catalyzed estradiol-3-glucuronidation demonstrated homotropic activation kinetics (S(50) = 22 microM, Hill coefficient, n = 1.9) whereas estradiol-17-glucuronidation (catalyzed by other UGT enzymes) followed Michaelis-Menten kinetics (K(m) = 7 microM). Estradiol 105-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 12386134-2 2002 In confirmation of previous work using alamethicin-treated HLM pooled from four livers, UGT1A1-catalyzed estradiol-3-glucuronidation demonstrated homotropic activation kinetics (S(50) = 22 microM, Hill coefficient, n = 1.9) whereas estradiol-17-glucuronidation (catalyzed by other UGT enzymes) followed Michaelis-Menten kinetics (K(m) = 7 microM). Estradiol 232-241 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 12386134-4 2002 Although the classic UGT1A1 substrate bilirubin was a weak competitive inhibitor of estradiol-3-glucuronidation, the estrogens and anthraflavic acid activated or inhibited estradiol-3-glucuronidation dependent on substrate and effector concentrations. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 12386134-4 2002 Although the classic UGT1A1 substrate bilirubin was a weak competitive inhibitor of estradiol-3-glucuronidation, the estrogens and anthraflavic acid activated or inhibited estradiol-3-glucuronidation dependent on substrate and effector concentrations. Estradiol 84-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 21-27 12351142-7 2002 Glucuronidation of BZ and its analogues exhibited the following relative ranking of UDP-glucuronosyltransferase (UGT) metabolism: UGT1A9>UGT1A4>>UGT2B7>UGT1A6 approximately UGT1A1. benzidine 19-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 185-191 12439224-1 2002 UDP-glucuronosyltransferase 1A1 (UGT1A1) is a polymorphic enzyme responsible for the glucuronidation of structurally diverse drugs, non-drug xenobiotics and endogenous compounds (e.g. bilirubin). Bilirubin 184-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-31 12439224-1 2002 UDP-glucuronosyltransferase 1A1 (UGT1A1) is a polymorphic enzyme responsible for the glucuronidation of structurally diverse drugs, non-drug xenobiotics and endogenous compounds (e.g. bilirubin). Bilirubin 184-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 12439228-2 2002 The mean (SD) value of serum bilirubin in the subjects with G6PD deficiency and homozygous variation in UGT1A1 gene was 51.3 (17.8) micromol/l, which was significantly higher compared to that in the other five subgroups. Bilirubin 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 12357057-0 2002 Relationship between bilirubin UDP-glucuronosyl transferase 1A1 gene and neonatal hyperbilirubinemia. Bilirubin 21-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-63 12167566-13 2002 Alpha-DHA-G was formed in incubations of DHA with expressed UGT1A9 (K(m) 32 microM, V(max) 8.9 pmol min(-1) mg(-1)) or UGT2B7 (K(m) 438 microM, V(max) 10.9 pmol mg(-1) min(-1)) but not with UGT1A1 or UGT1A6. artenimol 6-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 12181437-6 2002 Determination of intrinsic clearances predicts that hepatic UGT1A1 and UGT1A9 and the extrahepatic UGT1A7 are major components in SN-38-G formation, whereas a minor role is suggested for UGT1A6, UGT1A8, and UGT1A10. Irinotecan 130-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 12167566-13 2002 Alpha-DHA-G was formed in incubations of DHA with expressed UGT1A9 (K(m) 32 microM, V(max) 8.9 pmol min(-1) mg(-1)) or UGT2B7 (K(m) 438 microM, V(max) 10.9 pmol mg(-1) min(-1)) but not with UGT1A1 or UGT1A6. alpha-dha-g 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 190-196 12151360-0 2002 Sulforaphane and its glutathione conjugate but not sulforaphane nitrile induce UDP-glucuronosyl transferase (UGT1A1) and glutathione transferase (GSTA1) in cultured cells. sulforaphane 0-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 12198827-5 2002 Bilirubin-uridinediphosphate-glucuronosyltransferase (UGT1A1) is the only enzyme involved in the conjugation of bilirubin. Bilirubin 112-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 12151360-0 2002 Sulforaphane and its glutathione conjugate but not sulforaphane nitrile induce UDP-glucuronosyl transferase (UGT1A1) and glutathione transferase (GSTA1) in cultured cells. Glutathione 21-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 12151360-2 2002 Using a high-throughput microtitre plate assay and TaqMan real time quantitative RT-PCR to measure mRNA, we show that sulforaphane and its glutathione conjugate, but not the nitrile, increased significantly (P < 0.05) both UGT1A1 and GSTA1 mRNA levels in HepG2 and HT29 cells. sulforaphane 118-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 226-232 12151360-5 2002 The induction of UGT1A1 and GSTA1 mRNA by sulforaphane was time and concentration dependent. sulforaphane 42-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 12101428-3 2002 We have constructed a recombinant adenovirus, Ad-hUGT1A1-CTLA4Ig that coexpresses human bilirubin-uridinediphosphoglucuronate glucuronosyltransferase (hUGT1A1) and soluble murine CTLA4Ig, both driven by CMV immediate-early promoters. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-56 12185559-3 2002 Our objective was to investigate whether the UGT2B7 H268Y and UGT1A1*28 polymorphisms contribute to the variability in morphine glucuronide-to-morphine plasma ratios among cancer patients undergoing analgesic therapy with morphine. Morphine 119-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 12185559-3 2002 Our objective was to investigate whether the UGT2B7 H268Y and UGT1A1*28 polymorphisms contribute to the variability in morphine glucuronide-to-morphine plasma ratios among cancer patients undergoing analgesic therapy with morphine. Glucuronides 128-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 12101428-3 2002 We have constructed a recombinant adenovirus, Ad-hUGT1A1-CTLA4Ig that coexpresses human bilirubin-uridinediphosphoglucuronate glucuronosyltransferase (hUGT1A1) and soluble murine CTLA4Ig, both driven by CMV immediate-early promoters. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 151-158 12101428-7 2002 A second injection of Ad-hUGT1A1-CTLA4Ig normalized serum bilirubin. Bilirubin 58-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-32 12101428-12 2002 In contrast, after Ad-hUGT1A1 (which expresses UGT1A1 alone) injection, serum bilirubin remained normal for only 4 weeks, and returned to preinjection levels by day 120. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 22-29 12153551-5 2002 In addition, CYP3A and UDPGT are catalysts of many reactions with endobiotics such as steroid hormones. Steroids 86-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-28 12018987-7 2002 Human UGT1A1, UGT1A8, and UGT1A9 were shown to be especially active in conjugation of both flavonoids, whereas UGT1A4 and UGT1A10 and the isoenzymes from the UGTB family, UGT2B7 and UGT2B15, were less efficient. Flavonoids 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-12 12078936-0 2002 Predicting the risk of sporadic elevated bilirubin levels and diagnosing Gilbert"s syndrome by genotyping UGT1A1*28 promoter polymorphism. Bilirubin 41-50 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 12078936-3 2002 The aim of this investigation was to evaluate the correlation of unspecific elevated bilirubin levels and the occurrence of GS with a described polymorphism in the uridine diphosphat glucuronosyltransferase 1A1 (UGT1A1) in a predominately Caucasian population. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-210 12078936-3 2002 The aim of this investigation was to evaluate the correlation of unspecific elevated bilirubin levels and the occurrence of GS with a described polymorphism in the uridine diphosphat glucuronosyltransferase 1A1 (UGT1A1) in a predominately Caucasian population. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 12078936-10 2002 Genotyping of the UGT1A1 promoter polymorphism is a cheap and unequivocal method for predicting elevated and fluctuating bilirubin levels. Bilirubin 121-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 12499798-6 2002 METHODS: Promoter regions of the gene for bilirubin UGT-1A1 in twelve patients with Gilbert"s syndrome and twenty healthy subjects (controls) were sequenced. Bilirubin 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-59 12105841-1 2002 BACKGROUND & AIMS: Coinheritance of the A(TA)7TAA promoter variant in the uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1) gene and glucose-6-phosphate dehydrogenase (G6PD) deficiency is crucial to hyperbilirubinemia in white male neonates. Adenosine Monophosphate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-129 12105841-1 2002 BACKGROUND & AIMS: Coinheritance of the A(TA)7TAA promoter variant in the uridine 5"-diphosphate-glucuronosyl transferase 1A1 (UGT1A1) gene and glucose-6-phosphate dehydrogenase (G6PD) deficiency is crucial to hyperbilirubinemia in white male neonates. Adenosine Monophosphate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 12051676-6 2002 Catalytic activity of UGT1A01 was determined with 7-hydroxy-4-(trifluoromethyl)-coumarin and more specific human UGT1A1 substrates (1-naphthol, beta-estradiol, 17 alpha-ethinylestradiol, and bilirubin). 1-naphthol 132-142 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 12019197-3 2002 UGT1A1 and 1A8 were found to catalyze the formation of both the 6-beta- and 4"-beta-glucuronides, whereas UGT1A10 formed only the 4"-beta-glucuronide. 6-beta- and 4"-beta-glucuronides 64-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-14 12019197-3 2002 UGT1A1 and 1A8 were found to catalyze the formation of both the 6-beta- and 4"-beta-glucuronides, whereas UGT1A10 formed only the 4"-beta-glucuronide. 4"-beta-glucuronide 76-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-14 12019197-5 2002 Kinetic parameters for raloxifene glucuronidation by expressed UGT1A1 could not be determined due to limited substrate solubility. Raloxifene Hydrochloride 23-33 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 12023536-8 2002 Among six human UGT isozymes tested, UGT1A1 and 1A3 were capable of catalyzing the glucuronidation of both GFZ and SVA. Gemfibrozil 107-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-51 12023536-8 2002 Among six human UGT isozymes tested, UGT1A1 and 1A3 were capable of catalyzing the glucuronidation of both GFZ and SVA. simvastatin hydroxyacid 115-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-51 11950788-0 2002 Induction of human UDP-glucuronosyltransferase UGT1A1 by flavonoids-structural requirements. Flavonoids 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-53 11950788-13 2002 Together, these results strongly suggest that the flavonoid induction of UGT1A1 is through a novel nonaryl hydrocarbon receptor-mediated mechanism. Flavonoids 50-59 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 11950779-7 2002 Studies with expressed UDP glucuronosyltransferases (UGTs) revealed that both UGT1A1 and UGT1A3 were capable of forming the glucuronide conjugates and the corresponding lactones for all three statins. Glucuronides 124-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 11950779-7 2002 Studies with expressed UDP glucuronosyltransferases (UGTs) revealed that both UGT1A1 and UGT1A3 were capable of forming the glucuronide conjugates and the corresponding lactones for all three statins. Lactones 169-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 11950788-1 2002 Recent studies in our laboratory in the human hepatic and intestinal cell lines Hep G2 and Caco-2 have demonstrated induction of UGT1A1 by the flavonoid chrysin (5,7-dihydroxyflavone) using catalytic activity assays and Western and Northern blotting. Flavonoids 143-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 11950788-1 2002 Recent studies in our laboratory in the human hepatic and intestinal cell lines Hep G2 and Caco-2 have demonstrated induction of UGT1A1 by the flavonoid chrysin (5,7-dihydroxyflavone) using catalytic activity assays and Western and Northern blotting. chrysin 153-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 11950788-1 2002 Recent studies in our laboratory in the human hepatic and intestinal cell lines Hep G2 and Caco-2 have demonstrated induction of UGT1A1 by the flavonoid chrysin (5,7-dihydroxyflavone) using catalytic activity assays and Western and Northern blotting. chrysin 162-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 129-135 11950788-2 2002 In the present study, we examined which features of the flavonoid structures were associated with induction of UGT1A1 and whether common drug-metabolizing enzyme inducers also produce this induction. Flavonoids 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 11990381-0 2002 UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity. Irinotecan 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 11983459-0 2002 Crigler-Najjar syndrome type II in a caucasian patient resulting from two mutations in the bilirubin uridine 5"-diphosphate-glucuronosyltransferase (UGT1A1) gene. Bilirubin 91-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 11956667-1 2002 OBJECTIVE: We investigated how the conserved mutation (Y486D) changed the kinetic parameters of uridine diphosphate glucuronosyltransferase 1A1 and 1A6 (UGT1A1 and 1A6) for 2-amino-5-nitro-4-trifluoromethylphenol, which is a major metabolite of flutamide, a nonsteroidal antiandrogenic agent. nitro-4-trifluoromethylphenol 183-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-167 11956667-1 2002 OBJECTIVE: We investigated how the conserved mutation (Y486D) changed the kinetic parameters of uridine diphosphate glucuronosyltransferase 1A1 and 1A6 (UGT1A1 and 1A6) for 2-amino-5-nitro-4-trifluoromethylphenol, which is a major metabolite of flutamide, a nonsteroidal antiandrogenic agent. Flutamide 245-254 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-167 11956667-5 2002 The Michaelis constant (K(M)) for the aglycone of the wild-type UGT1A1 was double that of the mutant, but the K(M) for UDP-GA of the wild-type UGT1A1 was not significantly different from that of the mutant. CHEBI:166892 38-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 11956667-5 2002 The Michaelis constant (K(M)) for the aglycone of the wild-type UGT1A1 was double that of the mutant, but the K(M) for UDP-GA of the wild-type UGT1A1 was not significantly different from that of the mutant. Uridine Diphosphate 119-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 64-70 11956667-5 2002 The Michaelis constant (K(M)) for the aglycone of the wild-type UGT1A1 was double that of the mutant, but the K(M) for UDP-GA of the wild-type UGT1A1 was not significantly different from that of the mutant. Uridine Diphosphate 119-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 143-149 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 156-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 11990381-4 2002 Here we report the results from a prospective clinical pharmacogenetic study to determine the significance of UGT1A1*28 polymorphism on irinotecan disposition and toxicity in patients with cancer. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11990381-10 2002 The results suggest that screening for UGT1A1*28 polymorphism may identify patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan induced gastrointestinal and bone marrow toxicity. Irinotecan 95-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 11990381-10 2002 The results suggest that screening for UGT1A1*28 polymorphism may identify patients with lower SN-38 glucuronidation rates and greater susceptibility to irinotecan induced gastrointestinal and bone marrow toxicity. Irinotecan 153-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 11956667-1 2002 OBJECTIVE: We investigated how the conserved mutation (Y486D) changed the kinetic parameters of uridine diphosphate glucuronosyltransferase 1A1 and 1A6 (UGT1A1 and 1A6) for 2-amino-5-nitro-4-trifluoromethylphenol, which is a major metabolite of flutamide, a nonsteroidal antiandrogenic agent. Ganciclovir 173-181 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-167 11915038-0 2002 Hemolysis and bilirubin conjugation in association with UDP-glucuronosyltransferase 1A1 promoter polymorphism. Bilirubin 14-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-87 11906189-1 2002 The UDP-glucuronosyltransferase UGT1A1 plays a critical role in the detoxification of potentially neurotoxic bilirubin by conjugating it with glucuronic acid. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 11906189-1 2002 The UDP-glucuronosyltransferase UGT1A1 plays a critical role in the detoxification of potentially neurotoxic bilirubin by conjugating it with glucuronic acid. Glucuronic Acid 142-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 11906189-2 2002 We identified a polymorphism that results in a T to G substitution at nucleotide number -3263 of the phenobarbital-responsive enhancer module of the UGT1A1 gene, thereby significantly decreasing transcriptional activity as indicated by the luciferase-reporter assay. Phenobarbital 101-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 11906189-5 2002 Plasma total bilirubin levels in these double heterozygotes were significantly higher than those in control subjects carrying one or other of these mutations singly, indicating that compound heterozygous mutations may result in more strongly reduced UGT1A1 activity. Bilirubin 13-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 250-256 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-4 2002 Interindividual variation of sensitivity to irinotecan is related to large variations of biotransformation of the active metabolite SN-38, some of which would be caused by genetic polymorphism of UGT1A1, an isozyme responsible for the SN-38 glucuronidation. Irinotecan 235-240 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-202 11805731-9 2002 The 4 subjects carrying UGT1A1*28 allele had values of the AUC(SN-38)/AUC(SN-38G) above the 75th percentile of the total population, suggesting a potential pharmacogenetic/pharmacokinetic relationship. Irinotecan 63-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 11805731-9 2002 The 4 subjects carrying UGT1A1*28 allele had values of the AUC(SN-38)/AUC(SN-38G) above the 75th percentile of the total population, suggesting a potential pharmacogenetic/pharmacokinetic relationship. Irinotecan 74-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 11848303-8 2002 These results suggest that Kupffer cells play a major role in the mediation of ethanol-stimulated induction of UGT1A1 in liver parenchymal cells. Ethanol 79-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 11868392-6 2002 UGT1A1 conjugates bilirubin, and mutations of the gene cause hereditary unconjugated hyperbilirubinemias (Crigler-Najjar syndrome and Gilbert"s syndrome). Bilirubin 18-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 84-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 166-172 11990381-2 2002 We have previously demonstrated the role of UGT1A1 enzyme in the glucuronidation of SN-38 and a significant correlation between in vitro glucuronidation of SN-38 and UGT1A1 gene promoter polymorphism. Irinotecan 156-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 44-50 11425418-2 2001 To determine whether this has a genetic basis we compared the bilirubin levels and frequency of gallstones in patients with different alleles of the UGT*1 gene. Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-154 11714888-9 2001 APAP-UGT activities correlated best with propofol-UGT (r = 0.85; UGT1A9) and bilirubin-UGT (r = 0.66; UGT1A1) activities, but poorly with UGT1A6 protein (r = 0.30). Acetaminophen 0-4 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 11546782-0 2001 Homodimerization of human bilirubin-uridine-diphosphoglucuronate glucuronosyltransferase-1 (UGT1A1) and its functional implications. Bilirubin 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 92-98 11546782-1 2001 Genetic lesions of bilirubin-uridine-diphosphoglucuronate glucuronosyltransferase-1 (UGT1A1) completely or partially abolish hepatic bilirubin glucuronidation, causing Crigler-Najjar syndrome type 1 or 2, respectively. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 11546782-9 2001 Coexpression of mutagenized and wild-type hUGT1A1 in COS-7 cells showed that the mutant form markedly suppressed the catalytic activity of wild-type hUGT1A1. carbonyl sulfide 53-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-49 11546782-9 2001 Coexpression of mutagenized and wild-type hUGT1A1 in COS-7 cells showed that the mutant form markedly suppressed the catalytic activity of wild-type hUGT1A1. carbonyl sulfide 53-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-156 11695848-4 2001 The glucuronidation of SN-38 was catalysed by UGT1A1, UGT1A3, UGT1A6 and UGT1A9 as well as by liver microsomes. Irinotecan 23-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 11695848-5 2001 Among these UGT isoforms, UGT1A1 showed the highest activity of SN-38 glucuronidation at both low (1 microM) and high (200 microM) substrate concentrations. Irinotecan 64-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11695848-10 2001 The UGT isoforms involved in SN-38 glucuronidation could be classified into two types: low-Km types such as UGT1A1 and UGT1A9, and high-Km types such as UGT1A3 and UGT1A6, in terms of affinity toward substrate. Irinotecan 29-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 11695848-14 2001 The activity of SN-38 glucuronidation by liver microsomes and UGT1A1 was effectively inhibited by bilirubin. Irinotecan 16-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 11695848-14 2001 The activity of SN-38 glucuronidation by liver microsomes and UGT1A1 was effectively inhibited by bilirubin. Bilirubin 98-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 11556815-9 2001 The bilirubin conjugating UGT1A1 was mainly involved in the 3-O-glucuronidation of trans-resveratrol, whereas the phenol conjugating UGT1A6 activity was restricted to cis-resveratrol. Bilirubin 4-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11556815-9 2001 The bilirubin conjugating UGT1A1 was mainly involved in the 3-O-glucuronidation of trans-resveratrol, whereas the phenol conjugating UGT1A6 activity was restricted to cis-resveratrol. 3-o 60-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11556815-9 2001 The bilirubin conjugating UGT1A1 was mainly involved in the 3-O-glucuronidation of trans-resveratrol, whereas the phenol conjugating UGT1A6 activity was restricted to cis-resveratrol. Resveratrol 83-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11556815-9 2001 The bilirubin conjugating UGT1A1 was mainly involved in the 3-O-glucuronidation of trans-resveratrol, whereas the phenol conjugating UGT1A6 activity was restricted to cis-resveratrol. Resveratrol 167-182 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 11489791-5 2001 CPT-11 is subject to extensive metabolic conversion by various enzyme systems, including esterases to form SN-38, UGT1A1 mediating glucuronidation of SN-38, as well as CYP3A4, which forms several pharmacologically inactive oxidation products. Irinotecan 0-6 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 11489791-5 2001 CPT-11 is subject to extensive metabolic conversion by various enzyme systems, including esterases to form SN-38, UGT1A1 mediating glucuronidation of SN-38, as well as CYP3A4, which forms several pharmacologically inactive oxidation products. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 114-120 11408353-8 2001 Incubations containing PhIP as substrate formed direct PhIP-glucuronides in microsomes expressing UGT1A1, UGT1A4 and UGT1A9 but at levels averaging 53-fold lower than when N-hydroxy-PhIP was used as the substrate. phip-glucuronides 55-72 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 11343231-4 2001 The anti-UGT-positive sera from AIH type 2 patients revealed the strongest immunoreactivity against UGT1A1, the main UGT-isoform involved in the bilirubin glucuronidation. Bilirubin 145-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 11457662-11 2001 Using microsome preparation (from HK293 cell expressing monkey UGT1A01), the apparent K(m) values were 13, 5 and 6 microM for the conjugation of estradiol, 2-hydroxyestradiol and 2-hydroxyestrone, respectively, and were very similar to the values obtained with human UGT1A1. Estradiol 145-154 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 267-273 11457662-11 2001 Using microsome preparation (from HK293 cell expressing monkey UGT1A01), the apparent K(m) values were 13, 5 and 6 microM for the conjugation of estradiol, 2-hydroxyestradiol and 2-hydroxyestrone, respectively, and were very similar to the values obtained with human UGT1A1. 2-hydroxyestradiol 156-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 267-273 11457662-11 2001 Using microsome preparation (from HK293 cell expressing monkey UGT1A01), the apparent K(m) values were 13, 5 and 6 microM for the conjugation of estradiol, 2-hydroxyestradiol and 2-hydroxyestrone, respectively, and were very similar to the values obtained with human UGT1A1. 2-hydroxyestrone 179-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 267-273 11343253-0 2001 The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR. Phenobarbital 4-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 11302935-10 2001 We screened commercially available UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B7, and UGT2B15 for epirubicin glucuronidation. Epirubicin 99-109 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 35-41 11343253-1 2001 The UDP-glucuronosyltransferase, UGT1A1, is the critical enzyme responsible for detoxification of the potentially neurotoxic bilirubin by conjugating it with glucuronic acid. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 11343231-8 2001 (2) In vitro immunoinhibition experiments showed that glucuronidation of the anticancer drug flavopiridol by UGT1A1 was more strongly inhibited than its UGT1A9-mediated biotransformation. alvocidib 93-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 109-115 11343253-1 2001 The UDP-glucuronosyltransferase, UGT1A1, is the critical enzyme responsible for detoxification of the potentially neurotoxic bilirubin by conjugating it with glucuronic acid. Glucuronic Acid 158-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11343253-2 2001 For decades, phenobarbital (PB) treatment for hyperbilirubinemia has been known to increase expression of the UGT1A1 gene in liver. Phenobarbital 13-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11343253-2 2001 For decades, phenobarbital (PB) treatment for hyperbilirubinemia has been known to increase expression of the UGT1A1 gene in liver. Phenobarbital 28-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11343253-3 2001 We have now delineated the PB response activity to a 290-bp distal enhancer sequence (-3483/-3194) of the UGT1A1 gene. Phenobarbital 27-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 106-112 11259324-5 2001 Chemical inhibition experiments further prove the involvement of UGT1A1 and UGT1A9 in the formation of M1 and M2, as the UGT1A1 substrate bilirubin preferably inhibited M1 over M2 (K(i): 36 and 258 microM, respectively), whereas the UGT1A9 substrate propofol showed a more pronounced decrease in M2 but not in M1 formation (K(i): 47 and 142 microM, respectively). Bilirubin 138-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11259324-5 2001 Chemical inhibition experiments further prove the involvement of UGT1A1 and UGT1A9 in the formation of M1 and M2, as the UGT1A1 substrate bilirubin preferably inhibited M1 over M2 (K(i): 36 and 258 microM, respectively), whereas the UGT1A9 substrate propofol showed a more pronounced decrease in M2 but not in M1 formation (K(i): 47 and 142 microM, respectively). Bilirubin 138-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 121-127 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. 7-ethyl-10-hydroxycamptothecin glucuronide 78-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11259324-5 2001 Chemical inhibition experiments further prove the involvement of UGT1A1 and UGT1A9 in the formation of M1 and M2, as the UGT1A1 substrate bilirubin preferably inhibited M1 over M2 (K(i): 36 and 258 microM, respectively), whereas the UGT1A9 substrate propofol showed a more pronounced decrease in M2 but not in M1 formation (K(i): 47 and 142 microM, respectively). Propofol 250-258 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 65-71 11259359-7 2001 SN-38, the active metabolite of irinotecan, is glucuronidated to the inactive SN-38 glucuronide by UGT1A1, the isoform catalyzing bilirubin glucuronidation. Bilirubin 130-139 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 11259359-13 2001 A phenotyping procedure for UGT1A1 has not been identified and genotyping of the UGT1A1 promoter in patients receiving irinotecan may identify patients at increased risk of toxicity. Irinotecan 119-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 81-87 11259359-14 2001 A clinical trial at the University of Chicago is ongoing to demonstrate the predictive significance of UGT1A1 genotyping for irinotecan pharmacodynamics. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 11283142-2 2001 It is metabolized to TAS-103-glucuronide (TAS-103-G) predominantly by uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1). tas-103-glucuronide 21-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-125 11283142-2 2001 It is metabolized to TAS-103-glucuronide (TAS-103-G) predominantly by uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1). tas-103-glucuronide 21-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 11283142-2 2001 It is metabolized to TAS-103-glucuronide (TAS-103-G) predominantly by uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1). tas-103-g 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 70-125 11283142-2 2001 It is metabolized to TAS-103-glucuronide (TAS-103-G) predominantly by uridine diphosphate glucuronosyltransferase isoform 1A1 (UGT1A1). tas-103-g 42-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-133 11283142-4 2001 In addition, we evaluated the influence of UGT1A1 genotype on the pharmacokinetics and toxicity of TAS-103. 6-((2-(dimethylamino)ethyl)amino)-3-hydroxy-7H-indeno(2,1-c)quinolin-7-one 99-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 43-49 11442279-0 2001 Induction of UDP-glucuronosyltransferase UGT1A1 by the flavonoid chrysin in Caco-2 cells--potential role in carcinogen bioinactivation. Flavonoids 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 11177741-5 2001 Increased toxicity seen in patients treated with CPT-11 may be explained by polymorphism in the UGT1A1 gene, which is responsible for glucuronidation of the active metabolite of CPT-11. Irinotecan 49-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 11177741-5 2001 Increased toxicity seen in patients treated with CPT-11 may be explained by polymorphism in the UGT1A1 gene, which is responsible for glucuronidation of the active metabolite of CPT-11. Irinotecan 178-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 11179460-5 2001 Using microsome preparations from human embryonic kidney 293 cells stably expressing each of the 12 human and 11 monkey UGT enzymes cloned to date, the two EM-652-monoglucuronides were detected after incubation with microsomes containing human UGT1A1, UGT1A3, UGT1A8, UGT1A9, and monkey monUGT1A01, monUGT1A03, and monUGT1A09. em-652-monoglucuronides 156-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 244-250 11230743-0 2001 Hepatic uptake of organic anions affects the plasma bilirubin level in subjects with Gilbert"s syndrome mutations in UGT1A1. Bilirubin 52-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 117-123 11230743-1 2001 Although in Gilbert"s syndrome (GS), bilirubin glucuronidation is impaired due to an extra TA in the TATA box of the promoter of the gene for bilirubin UDP-glucuronosyltransferase 1 (UGT1A1), many GS homozygotes lack unconjugated hyperbilirubinemia. Bilirubin 37-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-189 11230743-8 2001 Within each GS subgroup with defined UGT1A1 mutations, the plasma bilirubin level is in part determined by the organic anion uptake rate, assessed by early plasma disappearance of low-dose BSP. Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11442279-6 2001 RESULTS: Western blot analysis showed that pretreatment of Caco-2 cells with 25 microM chrysin induced UGT1A1 without affecting the expression of UGTs 1A6, 1A9 and 2B7. chrysin 87-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 103-109 11442279-8 2001 Similarly, glucuronidation of quercetin was greatly increased in a UGT1A1-specific way. Quercetin 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 11442279-9 2001 The induction of UGT1A1 in the Caco-2 cells resulted in a 10-fold increase in the glucuronidation of N-hydroxy-PhIP. 2-hydroxyamino-1-methyl-6-phenylimidazo(4,5-b)pyridine 101-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 11442279-10 2001 CONCLUSION: Dietary flavonoid-mediated induction of intestinal UGT1A1 may be important for the glucuronidation and detoxification of colon and other carcinogens as well as for the presystemic metabolism of therapeutic drugs. Flavonoids 20-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 11783425-0 2001 UGT1A1 polymorphism predicts irinotecan toxicity: evolving proof. Irinotecan 29-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 11159800-4 2001 Intrinsic clearance values for human UGT1A1 and UGT2B7 substrates were an order of magnitude higher in DLM than in HLM (e.g., gemfibrozil: 31 microl/min/mg versus 3.0 microl/min/mg; ketoprofen: 2.4 microl/min/mg versus 0.2 microl/min/mg). Gemfibrozil 126-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11159800-4 2001 Intrinsic clearance values for human UGT1A1 and UGT2B7 substrates were an order of magnitude higher in DLM than in HLM (e.g., gemfibrozil: 31 microl/min/mg versus 3.0 microl/min/mg; ketoprofen: 2.4 microl/min/mg versus 0.2 microl/min/mg). Ketoprofen 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11174102-2 2001 Recently, the genetic basis of GS has been identified in caucasian populations : it is related to the insertion of a dinucleotide (TA) in the promoter region of the UGT1A1 gene. Dinucleoside Phosphates 117-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 11124229-2 2001 Four cloned and expressed human UDP-glucuronosyltransferases (UGT1A1, UGT1A6, UGT1A9, and UGT2B15) were used to screen a series of three potential drug substrates differing only in position of the phenol moiety. Phenol 197-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 62-68 11170257-0 2000 Bilirubin UDP-glucuronosyltransferase 1A1 gene polymorphisms: susceptibility to oxidative damage and cancer? Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-41 11156391-2 2000 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-131 11156391-2 2000 Irinotecan is metabolized to form active SN-38, which is further conjugated and detoxified by UDP-glucuronosyltransferase (UGT) 1A1 enzyme. Irinotecan 41-46 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-131 11156391-3 2000 Genetic polymorphisms of the UGT1A1 would affect an interindividual variation of the toxicity by irinotecan via the alternation of bioavailability of SN-38. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 11156391-3 2000 Genetic polymorphisms of the UGT1A1 would affect an interindividual variation of the toxicity by irinotecan via the alternation of bioavailability of SN-38. Irinotecan 150-155 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 11156391-4 2000 In this case-control study, retrospective review of clinical records and determination of UGT1A1 polymorphisms were performed to investigate whether a patient with the variant UGT1A1 genotypes would be at higher risk for severe toxicity by irinotecan. Irinotecan 240-250 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 11156391-9 2000 Multivariate analysis suggested that the genotype either heterozygous or homozygous for UGT1A1*28 would be a significant risk factor for severe toxicity by irinotecan (P < 0.001; odds ratio, 7.23; 95% confidence interval, 2.52-22.3). Irinotecan 156-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 88-94 11156391-13 2000 We suggest that determination of the UGT1A1 genotypes might be clinically useful for predicting severe toxicity by irinotecan in cancer patients. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11156391-14 2000 This research warrants a prospective trial to corroborate the usefulness of gene diagnosis of UGT1A1 polymorphisms prior tb irinotecan chemotherapy. Terbium 121-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 11156391-14 2000 This research warrants a prospective trial to corroborate the usefulness of gene diagnosis of UGT1A1 polymorphisms prior tb irinotecan chemotherapy. Irinotecan 124-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 11196269-2 2001 We analyzed the association of chemotherapy-induced hyperbilirubinemia with mutations of the bilirubin uridine-5"-diphosphate (UDP)-glucuronosyltransferase gene (UGT1A1) from two leukemic patients in whom chemotherapy resulted in a hyperbilirubinemic response. Bilirubin 57-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 11196269-2 2001 We analyzed the association of chemotherapy-induced hyperbilirubinemia with mutations of the bilirubin uridine-5"-diphosphate (UDP)-glucuronosyltransferase gene (UGT1A1) from two leukemic patients in whom chemotherapy resulted in a hyperbilirubinemic response. Uridine Diphosphate 103-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 11196269-2 2001 We analyzed the association of chemotherapy-induced hyperbilirubinemia with mutations of the bilirubin uridine-5"-diphosphate (UDP)-glucuronosyltransferase gene (UGT1A1) from two leukemic patients in whom chemotherapy resulted in a hyperbilirubinemic response. Uridine Diphosphate 127-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 11201174-1 2000 UDP-glucuronosyltransferases (UGTs) involved in troglitazone glucuronidation in rats and humans have been characterized to support the previous toxicity study on troglitazone in Gunn rats and to examine whether the UGT polymorphism or inhibition of bilirubin metabolism is related to the clinically reported rare cases of liver failure. Troglitazone 48-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-33 11201174-1 2000 UDP-glucuronosyltransferases (UGTs) involved in troglitazone glucuronidation in rats and humans have been characterized to support the previous toxicity study on troglitazone in Gunn rats and to examine whether the UGT polymorphism or inhibition of bilirubin metabolism is related to the clinically reported rare cases of liver failure. Troglitazone 162-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-33 11201174-1 2000 UDP-glucuronosyltransferases (UGTs) involved in troglitazone glucuronidation in rats and humans have been characterized to support the previous toxicity study on troglitazone in Gunn rats and to examine whether the UGT polymorphism or inhibition of bilirubin metabolism is related to the clinically reported rare cases of liver failure. Bilirubin 249-258 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-33 11201174-3 2000 In humans, contribution of UGT1A1 was estimated to be about 30% of the total troglitazone glucuronidation by UGTs, using human liver microsomes and recombinant UGTs. Troglitazone 77-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 11201174-4 2000 Other UGT1 and UGT2 enzymes seem to be responsible for the rest of the troglitazone glucuronidation in humans. Troglitazone 71-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 6-10 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. Troglitazone 37-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-23 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. Troglitazone 37-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-23 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. TROGLITAZONE GLUCURONIDE 151-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-23 11201174-5 2000 The multiplicity of UGTs involved in troglitazone glucuronidation in humans may allow even patients lacking bilirubin UGT (UGT1A1) activity to produce troglitazone glucuronide. TROGLITAZONE GLUCURONIDE 151-175 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 11170257-1 2000 The UDP-glucuronosyltransferase 1A1 (UGT1A1) gene product catalyzes the glucuronidation of serum bilirubin as part of normal heme catabolism. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-35 11170257-1 2000 The UDP-glucuronosyltransferase 1A1 (UGT1A1) gene product catalyzes the glucuronidation of serum bilirubin as part of normal heme catabolism. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11170257-1 2000 The UDP-glucuronosyltransferase 1A1 (UGT1A1) gene product catalyzes the glucuronidation of serum bilirubin as part of normal heme catabolism. Heme 125-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-35 11170257-1 2000 The UDP-glucuronosyltransferase 1A1 (UGT1A1) gene product catalyzes the glucuronidation of serum bilirubin as part of normal heme catabolism. Heme 125-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 11170257-5 2000 We hypothesize that the UGT1A1 TA repeats or other functional polymorphisms resulting in lower serum bilirubin levels may be predictive of genetic susceptibility to oxidative damage and cancer. Bilirubin 101-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 11038168-6 2000 However, UGT1A1 was the only UGT capable of catalyzing the formation of two glucuronides of the catecholic entacapone. Glucuronides 76-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 15-25 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 136-166 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11108431-6 2000 With regard to irinotecan, patients with Gilbert"s syndrome phenotype have reduced inactivation of the active topoisomerase I inhibitor 7-ethyl-10-hydroxycamptothecin (SN-38) caused by a mutation in the UDP-glucuronosyltransferase 1A1 gene promoter. Irinotecan 168-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 203-234 11038168-6 2000 However, UGT1A1 was the only UGT capable of catalyzing the formation of two glucuronides of the catecholic entacapone. entacapone 107-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 9-15 11013440-0 2000 Genetic lesions of bilirubin uridine-diphosphoglucuronate glucuronosyltransferase (UGT1A1) causing Crigler-Najjar and Gilbert syndromes: correlation of genotype to phenotype. Bilirubin 19-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 11061796-3 2000 During our study of defects of the bilirubin uridine diphosphate-glucuronosyltransferase gene (UGT1A1) in patients with hereditary unconjugated hyperbilirubinemia (Crigler-Najjar syndrome and Gilbert"s syndrome) and neonatal hyperbilirubinemia, we encountered a prolonged case associated with breastfeeding; after cessation of breastfeeding, the infant"s bilirubin level became normal. Bilirubin 35-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 95-101 11003624-1 2000 Recent research has shown that congenital nonhemolytic low grade hyperbilirubinemias in patients with Gilbert"s syndrome (GS) are linked to mutations in the TATA box upstream of the uridine 5"-diphosphoglucose glucuronosyltransferase (UGT1A1) gene leading to an impaired bilirubin glucuronidation. Bilirubin 70-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 235-241 11013440-2 2000 Bilirubin-UGT(1) (UGT1A1) is the only isoform that significantly contributes to the conjugation of bilirubin. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 11013440-2 2000 Bilirubin-UGT(1) (UGT1A1) is the only isoform that significantly contributes to the conjugation of bilirubin. Bilirubin 99-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 11013440-11 2000 Several structural mutations of UGT1A1, for example, a G71R substitution, have been reported to cause mild reduction of UGT activity toward bilirubin, resulting in mild hyperbilirubinemia, consistent with Gilbert syndrome. Bilirubin 140-149 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 11097345-4 2000 Polymorphisms in UGT1A1, the major bilirubin-glucuronidating form, often result in a decreased capacity to glucuronidate bilirubin, such as observed in Gilbert Syndrome and some forms of perinatal jaundice. Bilirubin 35-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 10999728-2 2000 In the liver, SN-38 is glucuronidated (SN-38G) by UGT1A1, which also conjugates bilirubin. Irinotecan 14-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 11097345-4 2000 Polymorphisms in UGT1A1, the major bilirubin-glucuronidating form, often result in a decreased capacity to glucuronidate bilirubin, such as observed in Gilbert Syndrome and some forms of perinatal jaundice. Bilirubin 121-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 10999728-2 2000 In the liver, SN-38 is glucuronidated (SN-38G) by UGT1A1, which also conjugates bilirubin. Irinotecan 39-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 10999728-2 2000 In the liver, SN-38 is glucuronidated (SN-38G) by UGT1A1, which also conjugates bilirubin. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 11182932-0 2000 Crigler-Najjar syndrome type II resulting from three different mutations in the bilirubin uridine 5"-diphosphate-glucuronosyltransferase (UGT1A1) gene. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 10950852-0 2000 Induction of UDP-glucuronosyltransferase UGT1A1 by the flavonoid chrysin in the human hepatoma cell line hep G2. Flavonoids 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 10950852-11 2000 9-fold induction of the glucuronidation of the oral contraceptive drug ethinylestradiol, two of the best known and specific UGT1A1 substrates, demonstrating the potential importance of this induction. Ethinyl Estradiol 71-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 124-130 10975608-1 2000 The activity of uridine-diphosphoglucuronosyl transferase 1 (UGT1) may influence the concentration of serum bilirubin. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-59 10946897-3 2000 The predominant thyroid hormone released from the thyroid gland, T4, and the inactive rT3 are glucuronidated by cloned expressed bilirubin UGT1A1 and also phenol UGT1A9. Bilirubin 129-138 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 10975608-1 2000 The activity of uridine-diphosphoglucuronosyl transferase 1 (UGT1) may influence the concentration of serum bilirubin. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 61-65 10975608-6 2000 Subjects with 6/7 or heterozygous variation within the coding region or compound heterozygous (plus one homozygous) variation had significantly higher bilirubin levels than those with wild UGT1A1 gene. Bilirubin 151-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 189-195 10975608-7 2000 When the 290 subjects were stratified into six groups according to their serum bilirubin concentrations, the bilirubin levels were correlated well to the frequencies of variant UGT1A1 gene. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 10975608-7 2000 When the 290 subjects were stratified into six groups according to their serum bilirubin concentrations, the bilirubin levels were correlated well to the frequencies of variant UGT1A1 gene. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 177-183 10975608-8 2000 Our results show that there is a strong association between UGT1A1 gene and bilirubin levels in healthy Taiwanese adults. Bilirubin 76-85 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 10506112-4 1999 UGT1A1, UGT1A4 and UGT1A9 each metabolized all of the aromatic amines. aromatic amines 54-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10821120-7 2000 When a beta-glucuronidase inhibitor such as saccharic acid 1,4-lactone, glycyrrhizin or 1-naphtyl-beta-D-glucuronide was added to the reaction mixture, the bilirubin conjugation activity of the human UGT1A1 was detected. 1-naphtyl-beta-d-glucuronide 88-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 200-206 10821120-7 2000 When a beta-glucuronidase inhibitor such as saccharic acid 1,4-lactone, glycyrrhizin or 1-naphtyl-beta-D-glucuronide was added to the reaction mixture, the bilirubin conjugation activity of the human UGT1A1 was detected. Bilirubin 156-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 200-206 10821120-8 2000 When geniposide was added to the reaction mixture, the bilirubin conjugation activity of UGT1A1 was not seen. geniposide 5-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 10821120-8 2000 When geniposide was added to the reaction mixture, the bilirubin conjugation activity of UGT1A1 was not seen. Bilirubin 55-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 11023036-7 2000 In the ER, bilirubin is conjugated by bilirubin uridine diphosphate (UDP)-glycosyltransferase (bilirubin UGT; UGT1A1) to form mono- and diglucuronides of bilirubin. mono- and diglucuronides 126-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11023036-7 2000 In the ER, bilirubin is conjugated by bilirubin uridine diphosphate (UDP)-glycosyltransferase (bilirubin UGT; UGT1A1) to form mono- and diglucuronides of bilirubin. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 10850406-1 2000 Gilbert"s syndrome, a genetic deficiency in bilirubin UDP-glucuronosyltransferase (UGT1A1), may dispose to increased toxicity of propafenone in poor metabolizers (PMs) of cytochrome P4502D6 because glucuronidation of propafenone is the major metabolic pathway for drug elimination in PMs. Propafenone 129-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10850406-1 2000 Gilbert"s syndrome, a genetic deficiency in bilirubin UDP-glucuronosyltransferase (UGT1A1), may dispose to increased toxicity of propafenone in poor metabolizers (PMs) of cytochrome P4502D6 because glucuronidation of propafenone is the major metabolic pathway for drug elimination in PMs. Propafenone 217-228 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 11023036-7 2000 In the ER, bilirubin is conjugated by bilirubin uridine diphosphate (UDP)-glycosyltransferase (bilirubin UGT; UGT1A1) to form mono- and diglucuronides of bilirubin. Bilirubin 11-20 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 11023036-7 2000 In the ER, bilirubin is conjugated by bilirubin uridine diphosphate (UDP)-glycosyltransferase (bilirubin UGT; UGT1A1) to form mono- and diglucuronides of bilirubin. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 110-116 10506112-9 1999 UGT1A1 demonstrated more extensive metabolism of the hydroxamic acid, N-hydroxy-N,N"-diacetylbenzidine, and the ring oxidation product, 3-OH-N,N"-diacetylbenzidine, than it did for the other six amines. Hydroxamic Acids 53-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10506112-9 1999 UGT1A1 demonstrated more extensive metabolism of the hydroxamic acid, N-hydroxy-N,N"-diacetylbenzidine, and the ring oxidation product, 3-OH-N,N"-diacetylbenzidine, than it did for the other six amines. N-hydroxy-N,N'-diacetylbenzidine 70-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10506112-9 1999 UGT1A1 demonstrated more extensive metabolism of the hydroxamic acid, N-hydroxy-N,N"-diacetylbenzidine, and the ring oxidation product, 3-OH-N,N"-diacetylbenzidine, than it did for the other six amines. 3-oh-n,n"-diacetylbenzidine 136-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10506112-9 1999 UGT1A1 demonstrated more extensive metabolism of the hydroxamic acid, N-hydroxy-N,N"-diacetylbenzidine, and the ring oxidation product, 3-OH-N,N"-diacetylbenzidine, than it did for the other six amines. Amines 195-201 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10506112-11 1999 The K(m) values for N-acetylbenzidine metabolism by UGT1A1 and UGT1A4 were 0.37 +/- 0.14 and 1.8 +/- 0.4 mM, respectively. N-acetylbenzidine 20-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-58 10506112-14 1999 Likewise, the O-glucuronide of 3-OH-benzidine was stable at pH 7.4, with 52% remaining at pH 5.5 after 24 h. These results suggest the following relative ranking of transferase metabolism: UGT1A9 > UGT1A4 > > UGT2B7 > UGT1A6 approximately UGT1A1. o-glucuronide 14-27 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 10506112-14 1999 Likewise, the O-glucuronide of 3-OH-benzidine was stable at pH 7.4, with 52% remaining at pH 5.5 after 24 h. These results suggest the following relative ranking of transferase metabolism: UGT1A9 > UGT1A4 > > UGT2B7 > UGT1A6 approximately UGT1A1. 3-oh-benzidine 31-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 251-257 10462540-1 1999 UDP-glucuronosyltransferase (UGT) 1A1 (UGT1A1) catalyzes the glucuronidation of bilirubin in liver. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-37 10530490-2 1999 Recent molecular genetic studies have determined that the clinical phenotype can be described by a dinucleotide polymorphism in the TATA box promoter of the bilirubin uridine diphosphate-glucuronosyltransferase (UGT-1A1) gene, most frequently (TA)7TAA, affecting up to 36% of Africans, but only 3% of Asians. Dinucleoside Phosphates 99-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-219 10462540-1 1999 UDP-glucuronosyltransferase (UGT) 1A1 (UGT1A1) catalyzes the glucuronidation of bilirubin in liver. Bilirubin 80-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 10471066-0 1999 UDP-glucuronosyltransferase (UGT1A1*28 and UGT1A6*2) polymorphisms in Caucasians and Asians: relationships to serum bilirubin concentrations. Bilirubin 116-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 10421657-5 1999 UGT1A1 protein level correlated strongly with both liver microsomal bilirubin UGT activity and liver UGT1A1 mRNA level (r(2) =.82 and.72, respectively). Bilirubin 68-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 10421657-8 1999 Hepatocytes isolated from the phenytoin-exposed donors exhibited marked declines in UGT1A1 mRNA levels during culturing. Phenytoin 30-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 10421657-9 1999 Induction studies using hepatocytes treated for 48 hours with phenobarbital (2 mmol/L), oltipraz (50 micromol/L), or 3-methylcholanthrene (2.5 micromol/L) revealed UGT1A1-inducing effects of phenobarbital, oltipraz, and, in particular, 3-methylcholanthrene. Methylcholanthrene 117-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 10381366-0 1999 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Irinotecan 19-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-119 10381366-0 1999 Glucuronidation of 7-ethyl-10-hydroxycamptothecin (SN-38) by the human UDP-glucuronosyltransferases encoded at the UGT1 locus. Irinotecan 51-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-119 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-61 10381366-3 1999 In this study, we examined the potential for each of the UGT1-encoded isoforms (UGT1A1 and UGT1A3 through UGT1A10) to glucuronidate SN-38. Irinotecan 132-137 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 10353933-2 1999 We studied whether the condition was associated with mutations in the gene for bilirubin uridine 5"-diphosphate-glucuronosyltransferase (UGT1A1), a key enzyme of bilirubin catabolism. Bilirubin 79-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 10353933-2 1999 We studied whether the condition was associated with mutations in the gene for bilirubin uridine 5"-diphosphate-glucuronosyltransferase (UGT1A1), a key enzyme of bilirubin catabolism. Bilirubin 162-171 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 10353933-3 1999 DESIGN: We analyzed the UGT1A1 gene in 25 Japanese neonates who had nonphysiologic hyperbilirubinemia (serum bilirubin >257 micromol/L) with no obvious cause. Bilirubin 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 10353933-7 1999 RESULTS: We found a polymorphism for UGT1A1 in exon 1; a G-->A transition at nucleotide 211 caused arginine to replace glycine at position 71 of corresponding protein product (G71R). Arginine 102-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 10353933-7 1999 RESULTS: We found a polymorphism for UGT1A1 in exon 1; a G-->A transition at nucleotide 211 caused arginine to replace glycine at position 71 of corresponding protein product (G71R). Glycine 122-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. thymine-adenine 95-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. thymine-adenine 95-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. Tantalum 112-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. Tantalum 112-114 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. Bilirubin 131-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 10471066-2 1999 Mutations in the promoter of the UGT1A1 gene (UGT1A1*28), resulting in 5, 7 or 8, instead of 6 thymine-adenine (TA) repeats, alter bilirubin conjugation. Bilirubin 131-140 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 10471066-7 1999 Within both ethnic groups, serum bilirubin increased with increased numbers of UGT1A1 promoter TA repeats (P = 0.0001). Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 79-85 10471066-8 1999 However, a strong ethnic group-by-UGT1A1 genotype interaction suggests that additional ethnic differences in bilirubin metabolism contribute to observed bilirubin concentrations. Bilirubin 109-118 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 10471066-8 1999 However, a strong ethnic group-by-UGT1A1 genotype interaction suggests that additional ethnic differences in bilirubin metabolism contribute to observed bilirubin concentrations. Bilirubin 153-162 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 10340924-0 1999 Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Irinotecan 43-48 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 10340924-0 1999 Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 10340924-0 1999 Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Bilirubin 87-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 143-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 150-180 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10340924-1 1999 BACKGROUND: Hepatic uridine diphosphate glucuronosyltransferase (UGT) isoform 1A1 (UGT1A1) is primarily responsible for the glucuronidation of SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of the anticancer agent irinotecan. Irinotecan 229-239 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 10340924-2 1999 UGT1A1, also catalyzing the glucuronidation of bilirubin, has been shown to have reduced activity in Gilbert"s syndrome. Bilirubin 47-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 9848110-3 1998 Three human liver UDP-glucuronosyltransferases, UGT2B7, UGT1A1, and UGT1A3, have been shown to catalyze the glucuronidation of catechol estrogens and lead to their enhanced elimination via urine or bile. catechol 127-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 10190918-2 1999 The aim of this study was to investigate whether a genetic mutation in the bilirubin UGT1A1 gene, which has been associated with Gilbert"s syndrome in adults, is a contributory factor in prolonged neonatal jaundice. Bilirubin 75-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 9841869-6 1998 The sudden interruption of maternal glucose supply signals the enhanced expression of UGT1A1, giving a novel explanation for the physiological induction of bilirubin glucuronidation in newborn infants. Glucose 36-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 9841869-6 1998 The sudden interruption of maternal glucose supply signals the enhanced expression of UGT1A1, giving a novel explanation for the physiological induction of bilirubin glucuronidation in newborn infants. Bilirubin 156-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 10091405-2 1999 In this paper we tested the hypothesis related to the variability of the glucuronidation bilirubin rate which depends on the configuration of the A(TA)nTAA motif of the UGT1*1 glucuronosyltransferase gene promoter. Bilirubin 89-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 169-175 10091406-3 1999 The presence of seven thymine adenine (TA) repeats reduces the efficiency of transcription of the UGT1A1 gene. thymine adenine 22-37 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 10091406-3 1999 The presence of seven thymine adenine (TA) repeats reduces the efficiency of transcription of the UGT1A1 gene. Tantalum 39-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 10091406-11 1999 This polymorphism, as well as the (TA)7 one, is associated with an increased level of bilirubin and a significant reduction of transcription activity of the UGT1A1 gene. Bilirubin 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 157-163 10547036-9 1999 The results indicate that one or more phenol-specific UDP-glucuronosyltransferase 1A isoforms are expressed at above normal levels in this tyrosinemic subject. Phenol 38-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-84 10547036-9 1999 The results indicate that one or more phenol-specific UDP-glucuronosyltransferase 1A isoforms are expressed at above normal levels in this tyrosinemic subject. tyrosinemic 139-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-84 9848110-8 1998 Flunitrazepam (FNZ), a competitive inhibitor of morphine glucuronidation in hepatic microsomes, competitively inhibited catechol estrogen glucuronidation catalyzed by UGT2B7(Y), UGT1A1, and UGT1A3. Flunitrazepam 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-184 9848110-8 1998 Flunitrazepam (FNZ), a competitive inhibitor of morphine glucuronidation in hepatic microsomes, competitively inhibited catechol estrogen glucuronidation catalyzed by UGT2B7(Y), UGT1A1, and UGT1A3. catechol 120-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-184 9848110-10 1998 FNZ competitively inhibited buprenorphine glucuronidation with UGT1A1 and UGT2B7 but had no inhibitory activity toward UGT1A3. Buprenorphine 28-41 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 9692996-0 1998 Required buried alpha-helical structure in the bilirubin UDP-glucuronosyltransferase, UGT1A1, contains a nonreplaceable phenylalanine. Phenylalanine 120-133 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 86-92 9692996-1 1998 A conserved hydrophobic region in the bilirubin-type UDP-glucuronosyltransferase isozyme was first uncovered as a consequence of a deleterious mutation in the UGT1A1 (HUG-Br1) isozyme of a Crigler-Najjar (CN) Type I patient. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 9692996-1 1998 A conserved hydrophobic region in the bilirubin-type UDP-glucuronosyltransferase isozyme was first uncovered as a consequence of a deleterious mutation in the UGT1A1 (HUG-Br1) isozyme of a Crigler-Najjar (CN) Type I patient. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-174 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Tyrosine 68-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Tyrosine 68-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 227-233 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Leucine 72-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Leucine 72-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 227-233 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Bilirubin 125-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 227-233 9692996-7 1998 The less hydrophobic buried helix in the phenolic-type UGT1A6 has a Tyr/Leu at position 170/171; this isoform glucuronidated bilirubin at 1/10 the level of that by UGT1A1 with a Km (bilirubin) of 25 microM compared to that for UGT1A1 of 5. Bilirubin 182-191 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 164-170 9726089-9 1998 SN-38 is eliminated mainly through conjugation by hepatic uridine glucuronosyltransferase (UGT*1.1), the same isoezyme responsible for glucuronidation of bilirubin. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-98 9789606-0 1998 UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 24-30 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-1 1998 BACKGROUND: Irinotecan (CPT-11) is metabolized by esterase to form a SN-38, which is further conjugated by UGT1A1. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 9789606-3 1998 We investigated whether there might be an inter-individual difference in pharmacokinetics of SN-38 and its glucuronide, depending on the genotypes of UGT1A1. Irinotecan 93-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 9726089-9 1998 SN-38 is eliminated mainly through conjugation by hepatic uridine glucuronosyltransferase (UGT*1.1), the same isoezyme responsible for glucuronidation of bilirubin. Bilirubin 154-163 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 91-98 9726089-10 1998 Grade 4 irinotecan-related toxicity (ie, neutropenia, diarrhea) has recently been reported in two patients with deficient UGT*1.1 activity. Irinotecan 8-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 9497253-2 1998 In all CN-1 patients studied, structural mutations in one of the five exons of the gene (UGT1A1) encoding the uridinediphosphoglucuronate glucuronosyltransferase (UGT) isoform bilirubin-UGT1 were implicated in the absence or inactivation of the enzyme. Bilirubin 176-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 9639672-7 1998 The UGT1A1*32 protein supported 0-10% normal bilirubin glucuronidation when expressed in COS-1 cells. Bilirubin 45-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 9639672-8 1998 The I294T coding defect seen at the second allele in SM (CN-II) generated the UGT1A1*33 mutant protein which supported 40-55% normal activity with a normal Km (2.5 microM) for bilirubin. Bilirubin 176-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 9653159-0 1998 Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Bilirubin 117-126 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 57-63 9653159-9 1998 We suggest that the unstable UGT1A1 polymorphism may serve to "fine-tune" the plasma bilirubin level within population groups, maintaining it at a high enough level to provide protection against oxidative damage, but at a level that is sufficiently low to prevent kernicterus in infants. Bilirubin 85-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 9497253-2 1998 In all CN-1 patients studied, structural mutations in one of the five exons of the gene (UGT1A1) encoding the uridinediphosphoglucuronate glucuronosyltransferase (UGT) isoform bilirubin-UGT1 were implicated in the absence or inactivation of the enzyme. Bilirubin 176-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-93 9497253-6 1998 Bilirubin-UGT1 mRNA is difficult to obtain, since it is expressed in the liver only. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-14 9466980-8 1998 Gunn rats and CN-I patients lacked SN-38 glucuronidating activity, indicating the role of UGT1 isoform in SN-38 glucuronidation. Irinotecan 106-111 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-94 9525311-9 1998 Although the mean level of bilirubin in homozygous Gunn rats 1-2 days after birth was already 14.5-fold higher than that of heterozygous siblings, treatment with Ad5/CMV/hUG-Br1 reduced plasma bilirubin to normal levels within 1 week. Bilirubin 27-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-177 9525311-9 1998 Although the mean level of bilirubin in homozygous Gunn rats 1-2 days after birth was already 14.5-fold higher than that of heterozygous siblings, treatment with Ad5/CMV/hUG-Br1 reduced plasma bilirubin to normal levels within 1 week. Bilirubin 193-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 170-177 9466980-10 1998 Intact SN-38 glucuronidation was observed only in HK293 cells transfected with the UGT1A1 isozyme. Irinotecan 7-12 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 83-89 9466980-11 1998 These results demonstrate that UGT1A1 is the isoform responsible for SN-38 glucuronidation. Irinotecan 69-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 9466980-12 1998 These findings indicate a genetic predisposition to the metabolism of irinotecan, suggesting that patients with low UGT1A1 activity, such as those with Gilbert"s syndrome, may be at an increased risk for irinotecan toxicity. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 9466980-12 1998 These findings indicate a genetic predisposition to the metabolism of irinotecan, suggesting that patients with low UGT1A1 activity, such as those with Gilbert"s syndrome, may be at an increased risk for irinotecan toxicity. Irinotecan 204-214 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 9028453-2 1997 This study utilizes this information to identify the bases of deficient bilirubin UDP-glucuronosyltransferase activity encoded by the UGT1A gene for the major bilirubin isozyme, HUG-Br1, in 3 Crigler-Najjar type I individuals and the genotype of an at-risk unborn sibling of one patient. Bilirubin 72-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 178-185 9224784-4 1997 SDS-PAGE and Western blot analysis of UGT1.1-transfected HK293 membrane proteins photolabeled with [11,12-3H]atRA revealed a protein of approximately 56 kDa that was labeled by [3H]atRA, detected by anti-pNP UGT antibody and not present in membranes from nontransfected HK293 cells. Sodium Dodecyl Sulfate 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 9224784-4 1997 SDS-PAGE and Western blot analysis of UGT1.1-transfected HK293 membrane proteins photolabeled with [11,12-3H]atRA revealed a protein of approximately 56 kDa that was labeled by [3H]atRA, detected by anti-pNP UGT antibody and not present in membranes from nontransfected HK293 cells. 11,12-3h 100-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 9224784-4 1997 SDS-PAGE and Western blot analysis of UGT1.1-transfected HK293 membrane proteins photolabeled with [11,12-3H]atRA revealed a protein of approximately 56 kDa that was labeled by [3H]atRA, detected by anti-pNP UGT antibody and not present in membranes from nontransfected HK293 cells. Tretinoin 109-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 9224784-4 1997 SDS-PAGE and Western blot analysis of UGT1.1-transfected HK293 membrane proteins photolabeled with [11,12-3H]atRA revealed a protein of approximately 56 kDa that was labeled by [3H]atRA, detected by anti-pNP UGT antibody and not present in membranes from nontransfected HK293 cells. Tritium 106-108 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 9224784-4 1997 SDS-PAGE and Western blot analysis of UGT1.1-transfected HK293 membrane proteins photolabeled with [11,12-3H]atRA revealed a protein of approximately 56 kDa that was labeled by [3H]atRA, detected by anti-pNP UGT antibody and not present in membranes from nontransfected HK293 cells. Tretinoin 181-185 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 9402181-2 1997 Preclinical studies showed that UGT*1.1 is the isozyme responsible for SN-38 glucuronidation. Irinotecan 71-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-39 9402181-3 1997 Patients with Gilbert"s syndrome have deficient UGT*1.1 activity, therefore may have an increased risk for related CPT-11 toxicity. Irinotecan 115-121 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-55 8869742-3 1996 Treatment of humans or hepatoma cell lines with drugs such as phenobarbital causes the induction of hepatic bilirubin UGT by increased transcription from the UGT1 gene. Phenobarbital 62-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-162 8968658-4 1996 Phenobarbital, phenytoin, primidone and carbamazepine are potent inducers of cytochrome P450 (CYP), epoxide hydrolase and uridine diphosphate glucuronosyltransferase (UDPGT) enzyme systems; oxcarbazepine is a weak inducer of CYP enzymes, probably acting on a few specific isoforms only. Phenobarbital 0-13 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-172 8968658-4 1996 Phenobarbital, phenytoin, primidone and carbamazepine are potent inducers of cytochrome P450 (CYP), epoxide hydrolase and uridine diphosphate glucuronosyltransferase (UDPGT) enzyme systems; oxcarbazepine is a weak inducer of CYP enzymes, probably acting on a few specific isoforms only. Phenytoin 15-24 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-172 8968658-4 1996 Phenobarbital, phenytoin, primidone and carbamazepine are potent inducers of cytochrome P450 (CYP), epoxide hydrolase and uridine diphosphate glucuronosyltransferase (UDPGT) enzyme systems; oxcarbazepine is a weak inducer of CYP enzymes, probably acting on a few specific isoforms only. Primidone 26-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-172 8968658-4 1996 Phenobarbital, phenytoin, primidone and carbamazepine are potent inducers of cytochrome P450 (CYP), epoxide hydrolase and uridine diphosphate glucuronosyltransferase (UDPGT) enzyme systems; oxcarbazepine is a weak inducer of CYP enzymes, probably acting on a few specific isoforms only. Carbamazepine 40-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-172 8806713-4 1996 The results show that both rat and human UGT1.1 catalyze the glucuronidation of opioids with a relative reactivity of buprenorphine > > nalorphine approximately naltrexone. Buprenorphine 118-131 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 8806713-4 1996 The results show that both rat and human UGT1.1 catalyze the glucuronidation of opioids with a relative reactivity of buprenorphine > > nalorphine approximately naltrexone. Nalorphine 142-152 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 8806713-4 1996 The results show that both rat and human UGT1.1 catalyze the glucuronidation of opioids with a relative reactivity of buprenorphine > > nalorphine approximately naltrexone. Naltrexone 167-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 8806713-5 1996 Comparison of glucuronidation activities in livers from Crigler-Najjar type 1 patients and normal patients indicates that UGT1.1 catalyzes at least 75% of buprenorphine conjugation in normal human liver. Buprenorphine 155-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 8806713-7 1996 It was found that both rat and human UGT1.1 exhibited comparable substrate specificities and efficiencies (Vmax/Km) of glucuronide formation for anthraquinones, coumarins, estrogens, flavonoids, and phenolic compounds. Glucuronides 119-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 8806713-7 1996 It was found that both rat and human UGT1.1 exhibited comparable substrate specificities and efficiencies (Vmax/Km) of glucuronide formation for anthraquinones, coumarins, estrogens, flavonoids, and phenolic compounds. Anthraquinones 145-159 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 8806713-7 1996 It was found that both rat and human UGT1.1 exhibited comparable substrate specificities and efficiencies (Vmax/Km) of glucuronide formation for anthraquinones, coumarins, estrogens, flavonoids, and phenolic compounds. Coumarins 161-170 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 8806713-7 1996 It was found that both rat and human UGT1.1 exhibited comparable substrate specificities and efficiencies (Vmax/Km) of glucuronide formation for anthraquinones, coumarins, estrogens, flavonoids, and phenolic compounds. Flavonoids 183-193 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 8806713-7 1996 It was found that both rat and human UGT1.1 exhibited comparable substrate specificities and efficiencies (Vmax/Km) of glucuronide formation for anthraquinones, coumarins, estrogens, flavonoids, and phenolic compounds. phenolic compounds 199-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 8619894-4 1996 Recent reports of gut transplantation to reverse the metabolic defect in Gunn rats raised further interest in the expression and distribution of human bilirubin (UDP-GTs (HUG Br 1 and HUG Br 2) in the human alimentary tract. Bilirubin 151-160 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 171-192 8869742-3 1996 Treatment of humans or hepatoma cell lines with drugs such as phenobarbital causes the induction of hepatic bilirubin UGT by increased transcription from the UGT1 gene. Bilirubin 108-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-162 8869742-4 1996 The upstream region of UGT1*1 (bilirubin UGT) was sequenced and found to contain consensus sequences for several transcriptional regulatory elements including a "BARBIE box". Bilirubin 31-40 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 8869742-10 1996 The regulation of the biliruibin UGT gene by drugs is not yet understood and it will be important to identify additional genetic elements possibly further than -2kb upstream of the UGT1*1 coding region, which regulate the expression of this gene. biliruibin 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-187 7852413-8 1995 Recessively inherited mutant alleles for the predominant bilirubin isozyme, the HUG-Br1 protein, substituted Arg for Gly at codon 276 (G276R) in exon 1 of UGT1A abolishing a conserved di-glycine. Bilirubin 57-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-87 7852413-8 1995 Recessively inherited mutant alleles for the predominant bilirubin isozyme, the HUG-Br1 protein, substituted Arg for Gly at codon 276 (G276R) in exon 1 of UGT1A abolishing a conserved di-glycine. Glycylglycine 184-194 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-87 7852413-9 1995 The mutant HUG-Br1-G276R protein expressed in COS-1 cells had no detectable bilirubin glucuronidating activity at either pH 7.6 or 6.4. carbonyl sulfide 46-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-18 9224500-5 1995 When jaundice results from inactivation of members of the UGT1 family, conjugation of certain phenols, such as the anesthetic propofol, or synthetic estrogens, such as ethinylestradiol, can also be impaired. Phenols 94-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-62 9224500-5 1995 When jaundice results from inactivation of members of the UGT1 family, conjugation of certain phenols, such as the anesthetic propofol, or synthetic estrogens, such as ethinylestradiol, can also be impaired. Propofol 126-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-62 9224500-5 1995 When jaundice results from inactivation of members of the UGT1 family, conjugation of certain phenols, such as the anesthetic propofol, or synthetic estrogens, such as ethinylestradiol, can also be impaired. Ethinyl Estradiol 168-184 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-62 1900006-0 1991 Induction of alkoxyresorufin O-dealkylases and UDP-glucuronosyl transferase by phenobarbital and 3-methylcholanthrene in primary cultures of porcine ciliary epithelial cells. Phenobarbital 79-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-75 7971731-5 1994 Morphine and tricyclic antidepressants are substrates of a liver microsomal uridine diphosphate glucuronyl transferase (UDPGT). Morphine 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-118 7971731-5 1994 Morphine and tricyclic antidepressants are substrates of a liver microsomal uridine diphosphate glucuronyl transferase (UDPGT). Morphine 0-8 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 120-125 7971731-7 1994 All drugs inhibited the morphine UDPGT. Morphine 24-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-38 7971731-9 1994 Lubrol PX activated the morphine-UDPGT four to five times. Polidocanol 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-38 7971731-9 1994 Lubrol PX activated the morphine-UDPGT four to five times. Morphine 24-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-38 8226884-8 1993 The code for the predominant bilirubin isozyme, the HUG-Br1 protein, is missing the phenylalanine codon at position 170 in exon 1 of UGT1A, abolishing a conserved diphenylalanine. Bilirubin 29-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-59 8226884-8 1993 The code for the predominant bilirubin isozyme, the HUG-Br1 protein, is missing the phenylalanine codon at position 170 in exon 1 of UGT1A, abolishing a conserved diphenylalanine. Phenylalanine 84-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-59 8226884-8 1993 The code for the predominant bilirubin isozyme, the HUG-Br1 protein, is missing the phenylalanine codon at position 170 in exon 1 of UGT1A, abolishing a conserved diphenylalanine. diphenylalanine 163-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 52-59 8226884-9 1993 We demonstrate that, at the pH (7.6) routinely used for bilirubin glucuronidation studies, both the HUG-Br1 protein and human liver microsomes have approximately one-third the activity seen at the major pH optimum of 6.4 and at low ionic strength. Bilirubin 56-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-107 8467709-2 1993 The sub-family of UGTs that conjugate bilirubin and phenolic compounds with glucuronic acid has been termed UGT1A1. Bilirubin 38-47 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 8467709-2 1993 The sub-family of UGTs that conjugate bilirubin and phenolic compounds with glucuronic acid has been termed UGT1A1. Glucuronic Acid 76-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 108-114 8467709-5 1993 In the present study, we used the cDNA of UGT1A1*4, a bilirubin-conjugating isoform, to localize the UGT1A1 locus in the human genome. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 8467709-5 1993 In the present study, we used the cDNA of UGT1A1*4, a bilirubin-conjugating isoform, to localize the UGT1A1 locus in the human genome. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 1520294-6 1992 In addition, 7,7,7-triphenylheptyl-UDP (0.25-0.50 mM) almost inhibited glucuronidation of 1-naphthol and testosterone catalyzed by the recombinant rat liver UGT-2B1 and human liver UGT-1A1, whose cDNA has been expressed in V79 cells. 7,7,7-Triphenylheptyl UDP 13-38 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-188 1520294-6 1992 In addition, 7,7,7-triphenylheptyl-UDP (0.25-0.50 mM) almost inhibited glucuronidation of 1-naphthol and testosterone catalyzed by the recombinant rat liver UGT-2B1 and human liver UGT-1A1, whose cDNA has been expressed in V79 cells. 1-naphthol 90-100 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-188 1520294-6 1992 In addition, 7,7,7-triphenylheptyl-UDP (0.25-0.50 mM) almost inhibited glucuronidation of 1-naphthol and testosterone catalyzed by the recombinant rat liver UGT-2B1 and human liver UGT-1A1, whose cDNA has been expressed in V79 cells. Testosterone 105-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 181-188 1900006-0 1991 Induction of alkoxyresorufin O-dealkylases and UDP-glucuronosyl transferase by phenobarbital and 3-methylcholanthrene in primary cultures of porcine ciliary epithelial cells. Methylcholanthrene 97-117 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 47-75 1900006-7 1991 UDP-GT activity increased about 5-fold in PB-treated PE cells and about 4-fold in PB-treated NPE cells in 48 hr. Phenobarbital 42-44 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 1900006-7 1991 UDP-GT activity increased about 5-fold in PB-treated PE cells and about 4-fold in PB-treated NPE cells in 48 hr. Phenobarbital 82-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 1900006-9 1991 Induction by PB and MC of ER O-dealkylase, PR O-dealkylase and UDP-GT activities in ciliary NPE and PE cells was inhibited almost completely by 3.5 microM cyclohexamide and 40 nM actinomycin D. Phenobarbital 13-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 1900006-9 1991 Induction by PB and MC of ER O-dealkylase, PR O-dealkylase and UDP-GT activities in ciliary NPE and PE cells was inhibited almost completely by 3.5 microM cyclohexamide and 40 nM actinomycin D. Methylcholanthrene 20-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 1900006-9 1991 Induction by PB and MC of ER O-dealkylase, PR O-dealkylase and UDP-GT activities in ciliary NPE and PE cells was inhibited almost completely by 3.5 microM cyclohexamide and 40 nM actinomycin D. 4-[2-(3,5-dimethyl-2-oxocyclohexyl)-2-hydroxyethyl]piperidine-2,6-dione 155-168 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 1900006-9 1991 Induction by PB and MC of ER O-dealkylase, PR O-dealkylase and UDP-GT activities in ciliary NPE and PE cells was inhibited almost completely by 3.5 microM cyclohexamide and 40 nM actinomycin D. Dactinomycin 179-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33805415-3 2021 We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. Irinotecan 157-167 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 1898728-1 1991 We report the isolation and characterization of two human liver cDNA clones, HUG-Br1 and HUG-Br2; each encodes a UDP-glucuronosyltransferase enzyme which glucuronidates bilirubin IX alpha to form both the IX alpha C8 and IX alpha C12 monoconjugates and a diconjugate. Bilirubin 169-178 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 77-84 33805415-3 2021 We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. belinostat 169-179 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 33805415-3 2021 We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. pazopanib 181-190 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 33805415-3 2021 We summarized meta-analyses, genome-wide association studies, clinical trials, drug labels, and guidelines relating to the impact of UGT1A1 polymorphisms on irinotecan, belinostat, pazopanib, or nilotinib toxicities. nilotinib 195-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 133-139 33805415-4 2021 For irinotecan, UGT1A1*28 was significantly associated with neutropenia and diarrhea, particularly with doses >= 180 mg/m2, supporting the use of UGT1A1 to guide irinotecan prescribing. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 33805415-4 2021 For irinotecan, UGT1A1*28 was significantly associated with neutropenia and diarrhea, particularly with doses >= 180 mg/m2, supporting the use of UGT1A1 to guide irinotecan prescribing. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33805415-5 2021 The drug label for belinostat recommends a reduced starting dose of 750 mg/m2 for UGT1A1*28 homozygotes, though published studies supporting this recommendation are sparse. belinostat 19-29 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 82-88 33805415-6 2021 There was a correlation between UGT1A1 polymorphisms and pazopanib-induced hepatotoxicity, though further studies are needed to elucidate the role of UGT1A1-guided pazopanib dose adjustments. pazopanib 57-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 32-38 33805415-6 2021 There was a correlation between UGT1A1 polymorphisms and pazopanib-induced hepatotoxicity, though further studies are needed to elucidate the role of UGT1A1-guided pazopanib dose adjustments. pazopanib 164-173 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 33232579-0 2021 Drug-drug interaction of atazanavir on UGT1A1-mediated glucuronidation of molidustat in human. Atazanavir Sulfate 25-35 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 33777142-0 2021 UGT1A1 Polymorphism for Irinotecan Dose Escalation in Patients with BRAF-Mutated Metastatic Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI. Irinotecan 24-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33777142-0 2021 UGT1A1 Polymorphism for Irinotecan Dose Escalation in Patients with BRAF-Mutated Metastatic Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI. FOLFIRI regimen 150-157 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33777142-3 2021 In this study, we explored the effects and oncological outcomes of UGT1A1 polymorphism for irinotecan escalation in patients with BRAF-mutated mCRC. Irinotecan 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 33762934-0 2021 UGT1A1 rs4148323 A Allele is Associated With Increased 2-Hydroxy Atorvastatin Formation and Higher Death Risk in Chinese Patients With Coronary Artery Disease. 2-hydroxyatorvastatin 55-77 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 33762934-6 2021 The patients carrying A allele for the rs4148323 polymorphism (UGT1A1) showed an increase in 2-hydroxy ATV/ATV ratio (p = 1.69E-07, false discovery rate [FDR] = 8.66E-03) relative to the value in individuals without the variant allele. Atorvastatin 103-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33762934-6 2021 The patients carrying A allele for the rs4148323 polymorphism (UGT1A1) showed an increase in 2-hydroxy ATV/ATV ratio (p = 1.69E-07, false discovery rate [FDR] = 8.66E-03) relative to the value in individuals without the variant allele. Atorvastatin 107-110 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 33762934-9 2021 In conclusion, our results suggested that the UGT1A1 rs4148323 A allele was associated with increased 2-hydroxy ATV formation and was a significant death risk factor in Chinese patients with CAD. Atorvastatin 112-115 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 33232579-0 2021 Drug-drug interaction of atazanavir on UGT1A1-mediated glucuronidation of molidustat in human. molidustat 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 33232579-4 2021 Based on preclinical findings, glucuronidation of molidustat was predominantly mediated by the 5"-diphospho-glucuronosyltransferase (UGT) isoform UGT1A1. molidustat 50-60 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 146-152 33232579-5 2021 Therefore, atazanavir, which is a potent inhibitor of UGT1A1, was chosen for the evaluation of pharmacokinetics and EPO release following a single oral dose of 25 mg molidustat. Atazanavir Sulfate 11-21 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 22325916-0 2012 Impact of UGT1A1 gene variants on total bilirubin levels in Gilbert syndrome patients and in healthy subjects. Bilirubin 40-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33235483-7 2020 Prevalent UGT1A1 gene [TA]7TAA promoter allelic variant UGT1A1*28, characterized by an extra TA repeat, is associated with low transcriptional and reduced enzymatic effectiveness, decreased SN38 active irinotecan metabolite glucuronidation, vs wild-type UGT1A1*1 [A(TA)6TAA]. Irinotecan 202-212 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 56-62 33235483-11 2020 Implementation of up-front evaluation of the five validated DPYD variants and UGT1A1*28 in the multidisciplinary molecular tumor board, also including CRC genetic characterization, addresses potential treatments with fluoropyrimidines and irinotecan associations at proper doses and schedules, particularly for early CRC, MCRC patients fit for intensive regimens or unfit for conventional regimens, requiring treatment modulations, and also for patients who experience severe, unexpected toxicities. fluoropyrimidines 217-234 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 33235483-11 2020 Implementation of up-front evaluation of the five validated DPYD variants and UGT1A1*28 in the multidisciplinary molecular tumor board, also including CRC genetic characterization, addresses potential treatments with fluoropyrimidines and irinotecan associations at proper doses and schedules, particularly for early CRC, MCRC patients fit for intensive regimens or unfit for conventional regimens, requiring treatment modulations, and also for patients who experience severe, unexpected toxicities. Irinotecan 239-249 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 78-84 32796590-2 2020 Normally, unconjugated bilirubin enters hepatocytes through the uptake transporters organic anion transporting polypeptide (OATP) 1B1 and 1B3, undergoes glucuronidation by the Phase II enzyme UDP glucuronosyltransferase 1A1 (UGT1A1), and conjugated forms are excreted into the bile by the canalicular export pump multidrug resistance protein 2 (MRP2). Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-223 32796590-2 2020 Normally, unconjugated bilirubin enters hepatocytes through the uptake transporters organic anion transporting polypeptide (OATP) 1B1 and 1B3, undergoes glucuronidation by the Phase II enzyme UDP glucuronosyltransferase 1A1 (UGT1A1), and conjugated forms are excreted into the bile by the canalicular export pump multidrug resistance protein 2 (MRP2). Bilirubin 23-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 225-231 22325916-2 2012 This work investigated the effect of UGT1A1 variants on total bilirubin levels in Gilbert patients (n=45) and healthy controls (n=161). Bilirubin 62-71 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 37-43 22325916-12 2012 Alterations in the UGT1A1 coding region seem to be associated with increased bilirubin levels, and, therefore, with Gilbert syndrome. Bilirubin 77-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 34734679-6 2022 Genotypes NR1I2 rs2472677 C>T, NR1I2 rs6785049 G>A, SLCO1B1 rs4363657 T>C, SLCO1A2 rs4762699 T>C, and UGT1A1 rs4148323 G>A contributed to individual variation in rocuronium. Rocuronium 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 102-108 34910929-8 2022 For UGT, negligible to moderate inhibition by vicagrel was observed with IC50 values of >50.0, >50.0, 28.2, 8.7, >50.0 and 28.2 muM for UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9 and UGT2B7, respectively. methyl 2-(2-acetoxy-6,7-dihydrothieno(3,2-c)pyridin-5(4H)-yl)-2-(2-chlorophenyl)acetate 46-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 136-142 34734679-9 2022 Genetic variations in NR1I2 rs2472677, NR1I2 rs6785049, SLCO1B1 rs4363657, SLCO1A2 rs4762699, and UGT1A1 rs4148323 were related to extensive interindividual variability in the clinical duration and total clinical action time of rocuronium. Rocuronium 228-238 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 98-104 34625956-5 2022 We used a common variant in UGT1A1 (rs6742078) associated with an 26% increase in bilirubin levels in the genetic studies. Bilirubin 82-91 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 34888851-4 2022 Letermovir is a substrate of UGT1A1/3, P-gp, and OATP1B, with its clearance primarily mediated by OATP1B. letermovir 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-37 34931017-4 2021 The most active compound 6, a lathyrane diterpenoid (5/11/3 ring system), dose-dependently activated hPXR with a high selectivity, and significantly upregulated the expression of hPXR downstream genes CYP3A4 and UGT1A1. lathyrane diterpenoid 30-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 212-218 34943103-2 2021 The aim of our study was to assess the role of bilirubin, and the heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variants, which are involved in bilirubin homeostasis, in the NAFLD development in adult patients. Bilirubin 95-104 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 34943103-2 2021 The aim of our study was to assess the role of bilirubin, and the heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variants, which are involved in bilirubin homeostasis, in the NAFLD development in adult patients. Bilirubin 189-198 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 135-141 34943103-5 2021 The (GT)n/(TA)n dinucleotide variations in HMOX1/UGT1A1 gene promoters, respectively, were analyzed by fragment analysis. Nitrogen 8-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 34943103-5 2021 The (GT)n/(TA)n dinucleotide variations in HMOX1/UGT1A1 gene promoters, respectively, were analyzed by fragment analysis. (ta)n dinucleotide 10-28 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 34697081-7 2022 When exposed to a low dose of CPT-11 (10 mg/kg), hUGT1A1HEP mice displayed greater intestinal inflammatory (IL-1beta and IL-6) insult in addition to p53-triggered apoptotic responses. Irinotecan 30-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-56 34697081-9 2022 When we examined the induction of ER stress in organoids with thapsigargin (TGN), an inhibitor of sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), apoptosis and the caspase surge that occurred in hUGT1A1HEP mice were blocked in hUGT1A1GI organoids. Thapsigargin 62-74 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 196-203 34895177-2 2021 OBJECTIVE: To investigate the association between neonatal hyperbilirubinemia and co-inheritance of G6PD deficiency and 211 G to A variation of UGT1A1 in Chaozhou city of eastern Guangdong province, the effects of G6PD deficiency and UGT1A1 gene variant on the bilirubin level were determined in neonates with hyperbilirubinemia. Bilirubin 261-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 144-150 34895177-2 2021 OBJECTIVE: To investigate the association between neonatal hyperbilirubinemia and co-inheritance of G6PD deficiency and 211 G to A variation of UGT1A1 in Chaozhou city of eastern Guangdong province, the effects of G6PD deficiency and UGT1A1 gene variant on the bilirubin level were determined in neonates with hyperbilirubinemia. Bilirubin 261-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 234-240 34847314-4 2021 Additional, well-planned trials of the UGT1A1 genotype-based approach to irinotecan therapy are predicted to reduce adverse drug events in people with the UGT1A1*28/*28 genotypes and improve treatment efficacy in the rest of the patients, which might be cost-effective. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 39-45 34478607-7 2021 Inhibition kinetic analyses demonstrated that MOA competitively inhibited UGT1A1-catalyzed NHPH-O-glucuronidation in both hUGT1A1 and HLM, with Ki values of 0.52 muM and 1.22 muM, respectively. nhph-o 91-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 34478607-7 2021 Inhibition kinetic analyses demonstrated that MOA competitively inhibited UGT1A1-catalyzed NHPH-O-glucuronidation in both hUGT1A1 and HLM, with Ki values of 0.52 muM and 1.22 muM, respectively. nhph-o 91-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 34860573-0 2022 All You Need to Know About UGT1A1 Genetic Testing for Patients Treated With Irinotecan: A Practitioner-Friendly Guide. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-33 34860573-2 2022 Severe neutropenia and diarrhea are common dose-limiting toxicities of irinotecan-based therapy, and UGT1A1 polymorphisms are one of the major risk factors of these toxicities. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 101-107 34860573-3 2022 In 2005, the US Food and Drug Administration revised the drug label to indicate that patients with UGT1A1*28 homozygous genotype should receive a decreased dose of irinotecan. Irinotecan 164-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 34926449-7 2021 In this study, we demonstrate that arbutin has a protective effect on alpha-naphthylisothiocyanate-induced cholestasis via upregulation of the levels of FXR and downstream enzymes associated with bile acid homeostasis such as Bsep, Ntcp, and Sult2a1, as well as Ugt1a1. Arbutin 35-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 262-268 34320604-3 2021 This case report demonstrates in vivo the finding of previous in-vitro research suggesting that the UGT1A1 7/7 mutation most commonly associated with Gilbert"s syndrome may result in decreased clozapine excretion. Clozapine 193-202 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 34847314-4 2021 Additional, well-planned trials of the UGT1A1 genotype-based approach to irinotecan therapy are predicted to reduce adverse drug events in people with the UGT1A1*28/*28 genotypes and improve treatment efficacy in the rest of the patients, which might be cost-effective. Irinotecan 73-83 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 155-161 34536821-0 2021 Upregulation of UGT1A1 expression by ursolic acid and oleanolic acid via the inhibition of the PKC/NF-kappaB signaling pathway. ursolic acid 37-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 34807779-1 2022 UDP-glucuronyltransferase 1A1 (UGT1A1) is a member of the Phase II metabolic enzyme family and the only enzyme that can metabolize detoxified bilirubin. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-29 34807779-1 2022 UDP-glucuronyltransferase 1A1 (UGT1A1) is a member of the Phase II metabolic enzyme family and the only enzyme that can metabolize detoxified bilirubin. Bilirubin 142-151 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 34192355-6 2021 RESULTS: Our data indicated that dabrafenib had a broad inhibitory effect on 4-MU glucuronidation by inhibiting the activities of UGTs, especially on UGT1A1, UGT1A7, UGT1A8, and UGT1A9, and dabrafenib can increase the area under curve (AUC) of co-administered drug. dabrafenib 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 150-156 34192355-7 2021 CONCLUSIONS: Dabrafenib is a strong inhibitor of several UGTs and the co-administration of dabrafenib with drugs primarily metabolized by UGT1A1, 1A7, 1A8, or 1A9 may induce potential DDIs. dabrafenib 13-23 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 34192355-7 2021 CONCLUSIONS: Dabrafenib is a strong inhibitor of several UGTs and the co-administration of dabrafenib with drugs primarily metabolized by UGT1A1, 1A7, 1A8, or 1A9 may induce potential DDIs. dabrafenib 91-101 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 138-144 34278601-16 2021 Most PHSF patients have UGT1A1 deficiency, which may be the target of rifampicin. Rifampin 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 34794220-1 2021 Objective: To investigate the correlation between UGT1A1 polymorphisms and the irinotecan plus S-1 regimen-induced toxicities in Chinese advanced esophageal squamous cell carcinoma (ESCC) patients. Irinotecan 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-56 34794220-5 2021 Irinotecan plus S-1 regimen-induced toxicities of patients with different UGT1A1 polymorphisms were observed. Irinotecan 0-10 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 34327766-0 2021 Impact of UGT1A1 Genotype on the Efficacy and Safety of Irinotecan-based Chemotherapy in Metastatic Colorectal Cancer. Irinotecan 56-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 10-16 34327766-2 2021 We evaluated the associations between the UGT1A1 genotype linked to adverse events - caused by irinotecan - and the efficacy and safety of mXELIRI and FOLFIRI. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 34327766-2 2021 We evaluated the associations between the UGT1A1 genotype linked to adverse events - caused by irinotecan - and the efficacy and safety of mXELIRI and FOLFIRI. mxeliri 139-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 34327766-2 2021 We evaluated the associations between the UGT1A1 genotype linked to adverse events - caused by irinotecan - and the efficacy and safety of mXELIRI and FOLFIRI. FOLFIRI regimen 151-158 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 42-48 34866848-2 2021 In most cases, GS is associated with the UGT1A1*28 polymorphism of UGT1A1 gene coding the enzyme bilirubin uridine diphosphate glucuronosyltransferase (UGT-1A) which plays a key role in the bilirubin metabolism. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 34866848-2 2021 In most cases, GS is associated with the UGT1A1*28 polymorphism of UGT1A1 gene coding the enzyme bilirubin uridine diphosphate glucuronosyltransferase (UGT-1A) which plays a key role in the bilirubin metabolism. Bilirubin 97-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 34866848-2 2021 In most cases, GS is associated with the UGT1A1*28 polymorphism of UGT1A1 gene coding the enzyme bilirubin uridine diphosphate glucuronosyltransferase (UGT-1A) which plays a key role in the bilirubin metabolism. Bilirubin 190-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 41-47 34866848-2 2021 In most cases, GS is associated with the UGT1A1*28 polymorphism of UGT1A1 gene coding the enzyme bilirubin uridine diphosphate glucuronosyltransferase (UGT-1A) which plays a key role in the bilirubin metabolism. Bilirubin 190-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 67-73 34866848-7 2021 Conclusions: Despite the fact that the level of serum bilirubin increases with the rise in the number of additional TA dinucleotides in the UGT1A1 gene promoter tests of clinical manifestations only (jaundice, fatigue, sleep disturbances, nausea, belching, and so on) and increased bilirubin levels in patients with normal liver function do not allow unequivocally diagnose GS. Bilirubin 54-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 34866848-7 2021 Conclusions: Despite the fact that the level of serum bilirubin increases with the rise in the number of additional TA dinucleotides in the UGT1A1 gene promoter tests of clinical manifestations only (jaundice, fatigue, sleep disturbances, nausea, belching, and so on) and increased bilirubin levels in patients with normal liver function do not allow unequivocally diagnose GS. ta dinucleotides 116-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 140-146 34536821-0 2021 Upregulation of UGT1A1 expression by ursolic acid and oleanolic acid via the inhibition of the PKC/NF-kappaB signaling pathway. Oleanolic Acid 54-68 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 34536821-8 2021 Phorbol 12-myristate 13-acetate (PMA) and lipopolysaccharide (LPS), the agonists of PKC and NF-kappaB signaling, respectively, significantly inhibit hp65-mediated UGT1A1 luciferase activity. Tetradecanoylphorbol Acetate 0-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 34536821-8 2021 Phorbol 12-myristate 13-acetate (PMA) and lipopolysaccharide (LPS), the agonists of PKC and NF-kappaB signaling, respectively, significantly inhibit hp65-mediated UGT1A1 luciferase activity. Tetradecanoylphorbol Acetate 33-36 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 163-169 34536821-10 2021 PMA and LPS do not affect UGT1A1 activity in p65-silenced HepG2 cells; however, UA and OA mildly influence UGT1A1 expression in these cells. Tetradecanoylphorbol Acetate 0-3 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-113 34698602-3 2021 UGT1A1 catalyzed beta-estradiol 3-beta-D-glucuronide formation showed allosteric sigmoidal kinetics in all enzyme systems; while UGT1A3 catalyzed CDCA 24-acyl-beta-D-glucuronide formation exhibited Michaelis-Menten kinetics in HLM, substrate inhibition kinetics in HIM and rUGT systems. Estradiol 17-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34698602-3 2021 UGT1A1 catalyzed beta-estradiol 3-beta-D-glucuronide formation showed allosteric sigmoidal kinetics in all enzyme systems; while UGT1A3 catalyzed CDCA 24-acyl-beta-D-glucuronide formation exhibited Michaelis-Menten kinetics in HLM, substrate inhibition kinetics in HIM and rUGT systems. 3-beta-d-glucuronide 32-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34698602-3 2021 UGT1A1 catalyzed beta-estradiol 3-beta-D-glucuronide formation showed allosteric sigmoidal kinetics in all enzyme systems; while UGT1A3 catalyzed CDCA 24-acyl-beta-D-glucuronide formation exhibited Michaelis-Menten kinetics in HLM, substrate inhibition kinetics in HIM and rUGT systems. 24-acyl-beta-d-glucuronide 151-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34703007-6 2022 SN-38 clearance was influenced by rs887829 in UGT1A1, pre-treatment total bilirubin, and EGFR rare variant burden. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 46-52 34703007-7 2022 Within each UGT1A1 genotype group, elevated pre-treatment total bilirubin and/or presence of at least one rare variant in EGFR resulted in significantly lower SN-38 clearance. Irinotecan 159-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 34814402-1 2021 Gilbert"s syndrome is a kind of benign inherited disease of bilirubin binding disorder, mainly due to the homozygous polymorphism A(TA)7TAA in the promoter of the gene for uridine diphosphate -glucuronosyltransferase 1A1 (UGT1A1), which is a TA insertion into the promoter, designated as UGT1A1*28, with UGT activity reduction to 30% of the normal value. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-220 34814402-1 2021 Gilbert"s syndrome is a kind of benign inherited disease of bilirubin binding disorder, mainly due to the homozygous polymorphism A(TA)7TAA in the promoter of the gene for uridine diphosphate -glucuronosyltransferase 1A1 (UGT1A1), which is a TA insertion into the promoter, designated as UGT1A1*28, with UGT activity reduction to 30% of the normal value. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 34814402-1 2021 Gilbert"s syndrome is a kind of benign inherited disease of bilirubin binding disorder, mainly due to the homozygous polymorphism A(TA)7TAA in the promoter of the gene for uridine diphosphate -glucuronosyltransferase 1A1 (UGT1A1), which is a TA insertion into the promoter, designated as UGT1A1*28, with UGT activity reduction to 30% of the normal value. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 288-294 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. dolutegravir 165-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 34486919-4 2021 Furthermore, prospective trials have shown that patients harboring the UGT1A1 *1/*1 and *1/*28 genotypes can tolerate higher doses of irinotecan, which may in turn impact on a better outcome. Irinotecan 134-144 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 34486919-5 2021 Upfront UGT1A1 genotyping could therefore be a usefulness strategy in order to individualize irinotecan dosing, but consensus on the recommended dose based on the UGT1A1 genotype is still lacking. Irinotecan 93-103 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 8-14 34486919-8 2021 EXPERT OPINION: Implementation of UGT1A1*28 and UGT1A1*6 genotyping in clinical practice is a first step toward personalizing irinotecan therapy. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-40 34486919-8 2021 EXPERT OPINION: Implementation of UGT1A1*28 and UGT1A1*6 genotyping in clinical practice is a first step toward personalizing irinotecan therapy. Irinotecan 126-136 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 34721047-7 2021 Study with recombinant human UGTs suggested that multiple UGT isoforms including UGT1A9, UGT1A7, UGT1A3, UGT1A4, UGT1A1, UGT2B7 and UGT1A8 are involved in the conversion of ticagrelor to ticagrelor-O-glucuronide with UGT1A9 showing highest catalytic activity. Ticagrelor 173-183 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 34721047-7 2021 Study with recombinant human UGTs suggested that multiple UGT isoforms including UGT1A9, UGT1A7, UGT1A3, UGT1A4, UGT1A1, UGT2B7 and UGT1A8 are involved in the conversion of ticagrelor to ticagrelor-O-glucuronide with UGT1A9 showing highest catalytic activity. ticagrelor-o-glucuronide 187-211 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 113-119 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. dolutegravir 165-177 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 34528449-3 2021 Results: A novel cis-eQTL, rs28967009, was identified for UGT1A1, which is predicted to upregulate UGT1A1 expression thereby potentially affecting the metabolism of dolutegravir and irinotecan, which are extensively prescribed in SA for HIV and colorectal cancer treatment, respectively. Irinotecan 182-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 99-105 34528449-4 2021 Conclusion: As increased UGT1A1 expression could affect the clinical outcome of dolutegravir and irinotecan treatment by increasing drug clearance, patients with the rs28967009A variant may require increased drug doses to reach therapeutic levels or should be prescribed alternative drugs. dolutegravir 80-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 34528449-4 2021 Conclusion: As increased UGT1A1 expression could affect the clinical outcome of dolutegravir and irinotecan treatment by increasing drug clearance, patients with the rs28967009A variant may require increased drug doses to reach therapeutic levels or should be prescribed alternative drugs. Irinotecan 97-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 34573035-0 2021 Bilirubin Links HO-1 and UGT1A1*28 Gene Polymorphisms to Predict Cardiovascular Outcome in Patients Receiving Maintenance Hemodialysis. Bilirubin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 25-31 34575495-2 2021 Resveratrol enantiomers showed stereoselective inhibition of CYP2C9, CYP3A, and UGT1A1. Resveratrol 0-11 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 34575495-5 2021 Trans-resveratrol noncompetitively inhibited CYP3A and UGT1A1 activities with Ki values of 23.8 and 27.4 muM, respectively. Resveratrol 0-17 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 34575495-8 2021 This information helped the prediction of food drug interaction potentials between resveratrol and co-administered drugs which are mainly metabolized by UGT1A1, CYP1A2, CYP2C19, CYP2E1, and CYP3A. Resveratrol 83-94 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 34572750-9 2021 Nine patients with toxicity-associated pharmacogenomic variants were treated with a relevant medication: seven UGT1A1 intermediate metabolizers were treated with irinotecan, one intermediate DPYD metabolizer was treated with 5-fluorouracil, and one TPMT poor metabolizer was treated with mercaptopurine. Irinotecan 162-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 111-117 34572750-11 2021 One UGT1A1 heterozygote died after a single dose of irinotecan due to irinotecan-related adverse effects. Irinotecan 52-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 34572750-11 2021 One UGT1A1 heterozygote died after a single dose of irinotecan due to irinotecan-related adverse effects. Irinotecan 70-80 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 4-10 34573035-1 2021 Serum bilirubin levels, which are determined by a complex interplay of various enzymes, including heme oxygenase-1 (HO-1) and uridine diphosphate-glucuronosyl transferase (UGT1A1), may be protective against progression of cardiovascular disease (CVD) in hemodialysis patients. Bilirubin 6-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 172-178 34573035-3 2021 This retrospective study enrolled 1080 prevalent hemodialysis patients and the combined genetic polymorphisms of HO-1 and UGT1A1 on serum bilirubin were analyzed. Bilirubin 138-147 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-128 34909672-4 2021 The metabolism pathways of dolutegravir and bictegravir are similar, both including CYP3A and UGT1A1, and both agents are substrates to different drug transporters. dolutegravir 27-39 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 34575401-5 2021 Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. dolutegravir 51-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 34575401-5 2021 Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Raltegravir Potassium 68-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 34575401-5 2021 Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Raltegravir Potassium 68-79 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 34575401-5 2021 Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Atazanavir Sulfate 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 33-39 34575401-5 2021 Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Atazanavir Sulfate 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 85-91 34909672-4 2021 The metabolism pathways of dolutegravir and bictegravir are similar, both including CYP3A and UGT1A1, and both agents are substrates to different drug transporters. bictegravir 44-55 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 94-100 34117260-0 2021 A UGT1A1 variant is associated with serum total bilirubin levels, which are causal for hypertension in African-ancestry individuals. Bilirubin 48-57 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 2-8 34149888-0 2021 Inhibitory effects of quercetin and its major metabolite quercetin-3-O-beta-D-glucoside on human UDP-glucuronosyltransferase 1A isoforms by liquid chromatography-tandem mass spectrometry. Quercetin 22-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-127 34149888-0 2021 Inhibitory effects of quercetin and its major metabolite quercetin-3-O-beta-D-glucoside on human UDP-glucuronosyltransferase 1A isoforms by liquid chromatography-tandem mass spectrometry. isoquercitrin 57-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 97-127 34149888-6 2021 Preliminary screening experiments indicated that quercetin exhibited stronger inhibitory effects on UGT1A1, UGT1A3, UGT1A6 and UGT1A9 enzymes than Q3GA. Quercetin 49-58 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 34149888-8 2021 Quercetin exerted non-competitive inhibition on UGT1A1 and UGT1A6, with half maximal inhibitory concentration (IC50) values of 7.47 and 7.07 microM and inhibition kinetic parameter (Ki) values of 2.18 and 28.87 microM, respectively. Quercetin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 34149888-11 2021 In the present study, quercetin was a moderate inhibitor of UGT1A1 and UGT1A3, a weak inhibitor of UGT1A6, and a strong inhibitor on UGT1A9. Quercetin 22-31 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 60-66 34149888-12 2021 The results of the present study suggested potential HDIs that may occur following quercetin co-administration with drugs that are mainly metabolized by UGT1A1, UGT1A3 and UGT1A9 enzymes. Quercetin 83-92 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 153-159 34085382-7 2021 YZH and its two active ingredients, namely baicalin (BCL) and 4"-hydroxyacetophenone (4-HT), up-regulated the mRNA expression of AhR target genes ( CYP1A1 and UGT1A1) and increased nuclear protein levels of AhR in HepG2 cells. baicalin 43-51 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 34085382-7 2021 YZH and its two active ingredients, namely baicalin (BCL) and 4"-hydroxyacetophenone (4-HT), up-regulated the mRNA expression of AhR target genes ( CYP1A1 and UGT1A1) and increased nuclear protein levels of AhR in HepG2 cells. baicalin 53-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 34085382-7 2021 YZH and its two active ingredients, namely baicalin (BCL) and 4"-hydroxyacetophenone (4-HT), up-regulated the mRNA expression of AhR target genes ( CYP1A1 and UGT1A1) and increased nuclear protein levels of AhR in HepG2 cells. 4-hydroxyacetophenone 62-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 34085382-7 2021 YZH and its two active ingredients, namely baicalin (BCL) and 4"-hydroxyacetophenone (4-HT), up-regulated the mRNA expression of AhR target genes ( CYP1A1 and UGT1A1) and increased nuclear protein levels of AhR in HepG2 cells. 4-hydroxyacetophenone 86-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 159-165 34117260-5 2021 The SNP rs887829 (UGT1A1) was significantly associated with TBIL levels (effect allele (T) frequency = 0.49, beta (SE) = 0.59 (0.04), p = 9.13 x 10-54). tbil 60-64 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 34117260-9 2021 Our finding confirms that UGT1A1 influences bilirubin levels. Bilirubin 44-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 26-32 34093191-9 2021 Results: In ATV-treated patients, hyperbilirubinemia (total bilirubin >1.2 mg/dl) had the main incidence (61.11%), and moderate and severe hyperbilirubinemia (total bilirubin >1.9 mg/dl) were statistically associated with UGT1A1*28 in recessive and codominant inheritance models (OR = 16.33, p = 0.028 and OR = 10.82, p = 0.036, respectively). Atazanavir Sulfate 12-15 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 34074250-0 2021 UGT1A1 mutation association with increased bilirubin levels and severity of unconjugated hyperbilirubinemia in ABO incompatible newborns of China. Bilirubin 43-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34074250-2 2021 Uridine diphosphate glucuronosyltransferase isoenzyme (UGT1A1) glucuronidates bilirubin and converts the toxic form of bilirubin to its nontoxic form. Uridine Diphosphate 0-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 34074250-2 2021 Uridine diphosphate glucuronosyltransferase isoenzyme (UGT1A1) glucuronidates bilirubin and converts the toxic form of bilirubin to its nontoxic form. Bilirubin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 34074250-2 2021 Uridine diphosphate glucuronosyltransferase isoenzyme (UGT1A1) glucuronidates bilirubin and converts the toxic form of bilirubin to its nontoxic form. Bilirubin 119-128 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 34074250-6 2021 UGT1A1 c.211 G > A mutation (UGT1A1*6, p.Arg71Gly, rs4148323) was significantly associated with the increased bilirubin level in ABO HDNs, after adjusted by age, sex and feeding method (P = 0.019 for TBIL, P = 0.02 for IBIL). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34074250-6 2021 UGT1A1 c.211 G > A mutation (UGT1A1*6, p.Arg71Gly, rs4148323) was significantly associated with the increased bilirubin level in ABO HDNs, after adjusted by age, sex and feeding method (P = 0.019 for TBIL, P = 0.02 for IBIL). Bilirubin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 29-35 34074250-6 2021 UGT1A1 c.211 G > A mutation (UGT1A1*6, p.Arg71Gly, rs4148323) was significantly associated with the increased bilirubin level in ABO HDNs, after adjusted by age, sex and feeding method (P = 0.019 for TBIL, P = 0.02 for IBIL). tbil 200-204 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34074250-6 2021 UGT1A1 c.211 G > A mutation (UGT1A1*6, p.Arg71Gly, rs4148323) was significantly associated with the increased bilirubin level in ABO HDNs, after adjusted by age, sex and feeding method (P = 0.019 for TBIL, P = 0.02 for IBIL). ibil 219-223 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34093191-3 2021 UGT1A1*28 and CYP2B6 c.516G>T have been proposed to be related with higher toxicity by ATV and EFV, respectively. Atazanavir Sulfate 87-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34093191-3 2021 UGT1A1*28 and CYP2B6 c.516G>T have been proposed to be related with higher toxicity by ATV and EFV, respectively. efavirenz 95-98 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34093191-9 2021 Results: In ATV-treated patients, hyperbilirubinemia (total bilirubin >1.2 mg/dl) had the main incidence (61.11%), and moderate and severe hyperbilirubinemia (total bilirubin >1.9 mg/dl) were statistically associated with UGT1A1*28 in recessive and codominant inheritance models (OR = 16.33, p = 0.028 and OR = 10.82, p = 0.036, respectively). Bilirubin 165-174 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 222-228 34093191-13 2021 Conclusion: Our findings suggest that after treatment with ATV or EFV, UGT1A1*28 and CYP2B6 c.516G>T influence the appearance of moderate-to-severe hyperbilirubinemia and CNS toxicity, respectively. Atazanavir Sulfate 59-62 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 71-77 35512135-7 2022 Of nine polymorphisms previously associated with dolutegravir pharmacokinetics, the lowest P-value with AUCVAR was UGT1A1 rs887829 (P = 1.8 x 10-4), which was also associated with log10 bilirubin (P = 8.6 x 10-13). dolutegravir 49-61 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 34272713-9 2021 Examples of adaptive nontoxic changes in liver function, which may elevate direct (conjugated) and/or indirect (unconjugated) bilirubin above baseline levels, include reversible inhibition of UGT1A1-mediated bilirubin metabolism and OATP1B1-, OATP1B3-, or MRP2-mediated transport. Bilirubin 126-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 34272713-9 2021 Examples of adaptive nontoxic changes in liver function, which may elevate direct (conjugated) and/or indirect (unconjugated) bilirubin above baseline levels, include reversible inhibition of UGT1A1-mediated bilirubin metabolism and OATP1B1-, OATP1B3-, or MRP2-mediated transport. Bilirubin 208-217 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 192-198 35415893-2 2022 The aim of this clinical study was to evaluate the safety, efficacy and pharmacokinetics of a novel regimen comprised of metronomic oxaliplatin (O), chronomodulated capecitabine (X) and UGT1A1 genotype-guided dosing of irinotecan (IRI) (OXIRI) as well as its immunomodulatory effects. Irinotecan 219-229 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 186-192 35415893-7 2022 UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. Irinotecan 71-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35415893-7 2022 UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. Irinotecan 83-88 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35415893-7 2022 UGT1A1-genotype directed dosing resulted in similar exposure levels of irinotecan, SN-38 and SN-38G in all patients. 7-ethyl-10-hydroxycamptothecin beta-glucuronide 93-99 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35633386-7 2022 RESULTS: The multivariate regression model revealed that patients who carried one or more detrimental allelic variant (CYP2D6, CYP3A5, and UGT1A1) showed a tendency toward a higher galantamine adjusted Cpss (B = 34.559, 95% CI = 0.741-68.377, p value = 0.045). Galantamine 181-192 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 139-145 35633386-9 2022 UGT1A1 mutant alleles and non-genetic factors including concomitant use of statin drugs and higher education level can attenuate therapeutic outcomes of galantamine. Galantamine 153-164 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35633386-10 2022 CONCLUSION: Pharmacokinetic-related genes including CYP2D6*10 and UGT1A1 mutant alleles were significantly associated with galantamine adjusted Cpss and cognitive function. Galantamine 123-134 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 34609801-3 2021 It happens as a result of an error in UGT1A1 enzyme which can cause high unconjugated bilirubin levels. Bilirubin 86-95 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 38-44 35527687-2 2022 The expression of UGT1A1 is relatively managed by a polymorphic dinucleotide repeat inside the promoter TATA box consisting of 5-8 copies of a TA repeat. Dinucleoside Phosphates 64-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 35637361-1 2022 BACKGROUND: UGT1A1 polymorphisms should be considered when using irinotecan-containing regimens, especially in patients with a double-variant-type (DV), including homozygous for UGT1A1*28 and UGT1A1*6 and heterozygous for both UGT1A1*28 and UGT1A1*6. Irinotecan 65-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 35177165-1 2022 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility and toxicity of patients to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-47 35512135-7 2022 Of nine polymorphisms previously associated with dolutegravir pharmacokinetics, the lowest P-value with AUCVAR was UGT1A1 rs887829 (P = 1.8 x 10-4), which was also associated with log10 bilirubin (P = 8.6 x 10-13). Bilirubin 186-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 115-121 35177165-1 2022 Uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) polymorphism plays a crucial role in the increased susceptibility and toxicity of patients to irinotecan. Irinotecan 151-161 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 49-55 35506159-0 2022 Association of UGT1A1*6,*28 or ABCC2 c.3972C>T genetic polymorphisms with irinotecan induced toxicity in Asian cancer patients: Meta-analysis. Irinotecan 74-84 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 35177165-2 2022 This retrospective, observational study compared the clinical outcomes and adverse events (AEs) in RAS wild-type metastatic colorectal cancer (mCRC) patients treated with cetuximab or bevacizumab plus FOLFIRI with UGT1A1 genotyping and irinotecan dose escalation as the first-line therapy. Irinotecan 236-246 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 214-220 35177165-5 2022 All patients received irinotecan dose escalation based on UGT1A1 genotyping. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 58-64 35177165-10 2022 Our results revealed that patients with wild-type RAS mCRC, regardless of biologics, with UGT1A1 genotyping can tolerate escalated doses of irinotecan and potentially achieve a more favorable clinical outcome without significantly increased toxicity. Irinotecan 140-150 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 90-96 35506159-3 2022 It was aimed to assess the aggregated risk of neutropenia or diarrhea in Asian cancer patients taking irinotecan and inherited UGT1A1*6, UGT1A1*28 or ABCC2 c.3972C>T genetic variants. Irinotecan 102-112 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 137-143 35506159-9 2022 Both UGT1A1*6 and UGT1A1*28 genetic variants should be screened in Asian cancer patients to reduce substantially irinotecan induced severe toxicities. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 5-11 35506159-9 2022 Both UGT1A1*6 and UGT1A1*28 genetic variants should be screened in Asian cancer patients to reduce substantially irinotecan induced severe toxicities. Irinotecan 113-123 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 18-24 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). Bilirubin 200-209 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35501760-6 2022 To the combined of UGT1A1, total bilirubin levels in homozygous variant were higher significantly than heterozygous variant and wild type (P = 0.002, P = 0.003, respectively). Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 19-25 35233973-5 2022 We also assessed the impact of UGT1A1 gene polymorphisms, which are involved in irinotecan metabolism, on outcomes and toxicity. Irinotecan 80-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 31-37 35233973-7 2022 Six (15%) patients (UGT1A1 wild-type (n = 2) and heterozygous (n = 4)) showed objective responses, including partial response (n = 1), tumor shrinkage (n = 4), and improved findings on their MIBG scan (n = 1). 3-Iodobenzylguanidine 191-195 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-0 2022 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by the Popular Flavonoids Baicalein, Baicalin and Hyperoside is responsible for Herbs (Shuang-huang-lian) -Induced Jaundice. Bilirubin 36-45 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-0 2022 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by the Popular Flavonoids Baicalein, Baicalin and Hyperoside is responsible for Herbs (Shuang-huang-lian) -Induced Jaundice. Flavonoids 77-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). Flavonoids 233-242 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35241486-0 2022 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by the Popular Flavonoids Baicalein, Baicalin and Hyperoside is responsible for Herbs (Shuang-huang-lian) -Induced Jaundice. baicalein 88-97 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). baicalein 261-270 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35241486-0 2022 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by the Popular Flavonoids Baicalein, Baicalin and Hyperoside is responsible for Herbs (Shuang-huang-lian) -Induced Jaundice. baicalin 99-107 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). baicalin 272-280 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35241486-0 2022 Inhibition of Human UGT1A1-Mediated Bilirubin Glucuronidation by the Popular Flavonoids Baicalein, Baicalin and Hyperoside is responsible for Herbs (Shuang-huang-lian) -Induced Jaundice. hyperoside 112-122 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35241486-3 2022 The aim of this paper was to explore the mechanism of Shuang-huang-lian injections (SHL) and its major constituents-induced jaundice via inhibiting human UDP-glucuronosyltransferases1A1 (hUGT1A1)-mediated bilirubin glucuronidation. Bilirubin 205-214 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 187-194 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). hyperoside 282-292 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35241486-7 2022 Both inhibition kinetics assays and molecular docking simulations suggested that SHL, BAI, BA, and HYP significantly inhibited hUGT1A1-mediated bilirubin glucuronidation via a mixed-type inhibition. Bilirubin 144-153 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 127-134 35445479-6 2022 SN-38 is inactivated by UGT1A1 enzymes. Irinotecan 0-5 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 24-30 35241486-8 2022 Collectively, some naturally occurring flavonoids (BAI, BA and HYP) in SHL have been identified as the inhibitors against hUGT1A1-mediated bilirubin glucuronidation, which well-explains the bilirubin-related ADRs or malady triggered by SHL in clinical settings. Flavonoids 39-49 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 35241486-8 2022 Collectively, some naturally occurring flavonoids (BAI, BA and HYP) in SHL have been identified as the inhibitors against hUGT1A1-mediated bilirubin glucuronidation, which well-explains the bilirubin-related ADRs or malady triggered by SHL in clinical settings. Bilirubin 139-148 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 35241486-8 2022 Collectively, some naturally occurring flavonoids (BAI, BA and HYP) in SHL have been identified as the inhibitors against hUGT1A1-mediated bilirubin glucuronidation, which well-explains the bilirubin-related ADRs or malady triggered by SHL in clinical settings. Bilirubin 190-199 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 122-129 35241486-10 2022 As a commonly used herbal products rich in flavonoids, Shuang-huang-lian injections (SHL), easily lead to symptoms of liver injury (e.g., jaundice) owing to significant inhibition of hUGT1A1-mediated bilirubin glucuronidation by its flavonoid components (i.e., baicalein, baicalin, hyperoside). Flavonoids 43-53 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 183-190 35147853-0 2022 Prediction of Drug-Drug Interaction Between Dabrafenib and Irinotecan via UGT1A1-Mediated Glucuronidation. dabrafenib 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 35147853-0 2022 Prediction of Drug-Drug Interaction Between Dabrafenib and Irinotecan via UGT1A1-Mediated Glucuronidation. Irinotecan 59-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 74-80 35147853-4 2022 METHODS: Recombinant human uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and human liver microsomes (HLMs) were employed to catalyze the glucuronidation of SN-38 in vitro. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 27-74 35147853-4 2022 METHODS: Recombinant human uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and human liver microsomes (HLMs) were employed to catalyze the glucuronidation of SN-38 in vitro. Irinotecan 167-172 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 76-82 35147853-6 2022 RESULTS: Dabrafenib noncompetitively inhibited SN-38 glucuronidation in pooled HLMs and recombinant UGT1A1 with unbound inhibitor constant (Ki,u) values of 12.43 +- 0.28 and 3.89 +- 0.40 muM, respectively. dabrafenib 9-19 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 35147853-6 2022 RESULTS: Dabrafenib noncompetitively inhibited SN-38 glucuronidation in pooled HLMs and recombinant UGT1A1 with unbound inhibitor constant (Ki,u) values of 12.43 +- 0.28 and 3.89 +- 0.40 muM, respectively. Irinotecan 47-52 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106 35147853-8 2022 However, the ratios of intra-enterocyte concentration of dabrafenib to Ki,u ((I)gut/Ki,u) are 2.73 and 8.72 in HLMs and recombinant UGT1A1, respectively, indicating a high risk of intestinal DDI when dabrafenib was used in combination with irinotecan. dabrafenib 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 132-138 35147853-9 2022 CONCLUSION: Dabrafenib is a potent noncompetitive inhibitor of UGT1A1 and may bring potential risk of DDI when combined with irinotecan. dabrafenib 12-22 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 35147853-9 2022 CONCLUSION: Dabrafenib is a potent noncompetitive inhibitor of UGT1A1 and may bring potential risk of DDI when combined with irinotecan. Irinotecan 125-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 35149777-0 2022 UGT1A1 genetic variants are associated with increases in bilirubin levels in rheumatoid arthritis patients treated with sarilumab. Bilirubin 57-66 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35149777-5 2022 Variants in the UGT1A1 gene were strongly associated with maximum bilirubin elevations in sarilumab-treated patients (rs4148325; p = 2.88 x 10-41) but were not associated with aminotransferase elevations. Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 16-22 35149777-7 2022 These findings suggest that most bilirubin increases during sarilumab treatment are related to genetic variation in UGT1A1 rather than underlying liver injury. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 116-122 35498511-0 2022 Clinical Outcomes of Patients with Peritoneal Metastasis-Only Colorectal Cancer Treated with First-Line Bevacizumab and FOLFIRI Through Irinotecan Dose Escalation According to UGT1A1 Polymorphism: Compared to Liver Metastasis-Only, and Lung Metastasis-Only. FOLFIRI regimen 120-127 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 176-182 35498511-8 2022 All patients with PC-only CRC were treated with first-line bevacizumab and FOLFIRI, and the irinotecan dose escalation depended on UGT1A1 polymorphism. Irinotecan 92-102 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 131-137 35498511-13 2022 Conclusion: Our study revealed that in patients with PC-only mCRC treatment of first-line bevacizumab and FOLFIRI through irinotecan dose escalation according to UGT1A1 polymorphism could confer such patients with comparable outcomes to that of patients with LiM-only and LuM-only mCRC. FOLFIRI regimen 106-113 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 35498511-13 2022 Conclusion: Our study revealed that in patients with PC-only mCRC treatment of first-line bevacizumab and FOLFIRI through irinotecan dose escalation according to UGT1A1 polymorphism could confer such patients with comparable outcomes to that of patients with LiM-only and LuM-only mCRC. Irinotecan 122-132 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 162-168 35436954-2 2022 BACKGROUND: Several mutations of bilirubin uridine diphosphate-glucuronosyltransferase gene (UGT1A1) have been reported in patients with unconjugated hyperbilirubinemia. Bilirubin 33-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 93-99 35061524-0 2022 Precision Medicine: UGT1A1 Genotyping to Better Manage Irinotecan-Induced Toxicity. Irinotecan 55-65 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 20-26 35226656-2 2022 Mutations in the UGT1A1 gene can be associated with increased toxicity from irinotecan-based chemotherapy. Irinotecan 76-86 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 17-23 35063619-4 2022 Myricetin exhibited strong inhibitory effects against UGT1A1, 1A3, 1A6, 1A7, 1A10 (IC50 < 10 muM) with non-competitive inhibition type, while serving as a moderate inhibitor against UGT1A9 and 2B7 (IC50 range from 25 to 29 muM) with competitive and mixed inhibition type, respectively. myricetin 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 54-60 35063619-7 2022 The quantitative prediction in vivo of inhibition on gastrointestinal UGTs by myricetin showed that the inhibition against UGT1A1, 1A3, 1A6, 1A7, 1A9, 1A10 and 2B7 would likely occur with high risk. myricetin 78-87 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 123-129 35226656-5 2022 Genetic testing identified a mutation in the UGT1A1 gene associated with increased toxicity to irinotecan, and the EIA tests performed for evaluation of C. difficile toxins A and B showed repeatedly negative results. Irinotecan 95-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 34996412-1 2022 BACKGROUND: Pharmacogenetic (PGx) testing for germline variants in the DPYD and UGT1A1 genes can be used to guide fluoropyrimidine and irinotecan dosing, respectively. 2-fluoropyrimidine 114-130 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 34654938-3 2022 Patients with genotype UGT1A1*6 (exon 1, 211G > A) treated with the antineoplastic drug SN-38 have been reported to exhibit decreased glucuronide conjugation and increased incidence of intestinal toxicity and its severe side effects, including severe diarrhea. Irinotecan 88-93 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 34654938-3 2022 Patients with genotype UGT1A1*6 (exon 1, 211G > A) treated with the antineoplastic drug SN-38 have been reported to exhibit decreased glucuronide conjugation and increased incidence of intestinal toxicity and its severe side effects, including severe diarrhea. Glucuronides 134-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 23-29 34654938-7 2022 The CYP3A4 POR UGT1A1*6 KI-Caco-2 cells were sensitive to SN-38-induced intestinal toxicity. Irinotecan 58-63 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 15-21 35192750-12 2022 There was no significant difference in the concentration of ALT, AST, cholesterol or LDL between different genotypes.The number of TA-repeats of the UGT1A1 gene affects the increase of total bilirubin and its indirect fraction, including the cases of rare allelic variants (TA<=5, TA>=8), but not the activity of ALT and AST and the lipid profile. Cholesterol 70-81 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 35192750-12 2022 There was no significant difference in the concentration of ALT, AST, cholesterol or LDL between different genotypes.The number of TA-repeats of the UGT1A1 gene affects the increase of total bilirubin and its indirect fraction, including the cases of rare allelic variants (TA<=5, TA>=8), but not the activity of ALT and AST and the lipid profile. Bilirubin 191-200 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 149-155 35264954-5 2022 Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. biflavones 46-56 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-165 35264954-5 2022 Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. bilobetin 67-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-165 35264954-5 2022 Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. isoginkgetin 78-90 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-165 35264954-5 2022 Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. sciadopitysin 92-105 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-165 35264954-5 2022 Further investigations demonstrated that four biflavones including bilobetin, isoginkgetin, sciadopitysin and ginkgetin, are key constituents responsible for hUGT1A1 inhibition in living cells. ginkgetin 110-119 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 158-165 35264954-6 2022 These biflavones potently inhibit hUGT1A1 in both human liver microsomes (HLM) and living cells, with the IC50 values ranging from 0.075 to 0.41 muM in living cells. biflavones 6-16 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 34-41 35264954-7 2022 Inhibition kinetic analyses and docking simulations suggested that four tested biflavones potently inhibit hUGT1A1-catalyzed NHPN-O-glucuronidation in HLM via a mixed inhibition manner, showing the K i values ranging from 0.07 to 0.74 muM. biflavones 79-89 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-114 35264954-7 2022 Inhibition kinetic analyses and docking simulations suggested that four tested biflavones potently inhibit hUGT1A1-catalyzed NHPN-O-glucuronidation in HLM via a mixed inhibition manner, showing the K i values ranging from 0.07 to 0.74 muM. nhpn 125-129 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 107-114 35207692-1 2022 Current guidelines recommend pre-therapeutic UGT1A1 genotyping to guide irinotecan dosing, but the usefulness of this approach remains to be clarified. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 45-51 34998046-0 2022 UGT1A1 genotype-guided dosing of irinotecan: A prospective safety and cost analysis in poor metaboliser patients. Irinotecan 33-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 34998046-1 2022 AIM: To determine the safety, feasibility, pharmacokinetics, and cost of UGT1A1 genotype-guided dosing of irinotecan. Irinotecan 106-116 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 73-79 34998046-2 2022 PATIENTS AND METHODS: In this prospective, multicentre, non-randomised study, patients intended for treatment with irinotecan were pre-therapeutically genotyped for UGT1A1*28 and UGT1A1*93. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 165-171 34998046-2 2022 PATIENTS AND METHODS: In this prospective, multicentre, non-randomised study, patients intended for treatment with irinotecan were pre-therapeutically genotyped for UGT1A1*28 and UGT1A1*93. Irinotecan 115-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 179-185 34998046-9 2022 Systemic exposure of SN-38 of reduced dosing in UGT1A1 PMs was still slightly higher compared to a standard-dosed irinotecan patient cohort (difference: +32%). Irinotecan 21-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-54 34998046-11 2022 CONCLUSION: UGT1A1 genotype-guided dosing significantly reduces the incidence of febrile neutropenia in UGT1A1 PM patients treated with irinotecan, results in a therapeutically effective systemic drug exposure, and is cost-saving. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 12-18 34998046-11 2022 CONCLUSION: UGT1A1 genotype-guided dosing significantly reduces the incidence of febrile neutropenia in UGT1A1 PM patients treated with irinotecan, results in a therapeutically effective systemic drug exposure, and is cost-saving. Irinotecan 136-146 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 34998046-12 2022 Therefore, UGT1A1 genotype-guided dosing of irinotecan should be considered standard of care in order to improve individual patient safety. Irinotecan 44-54 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 11-17 35017453-2 2022 High levels of raltegravir and dolutegravir can potentially cause bilirubin toxicity as they compete for albumin binding and follow the same metabolic pathway through UGT1A1. Raltegravir Potassium 15-26 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 35017453-2 2022 High levels of raltegravir and dolutegravir can potentially cause bilirubin toxicity as they compete for albumin binding and follow the same metabolic pathway through UGT1A1. dolutegravir 31-43 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 35017453-2 2022 High levels of raltegravir and dolutegravir can potentially cause bilirubin toxicity as they compete for albumin binding and follow the same metabolic pathway through UGT1A1. Bilirubin 66-75 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 167-173 35281975-6 2022 Results: Results showed that the processes of albumin-bound bilirubin transfer to the hepatocytes, hepatic uptake, and storage via ligandin, hepatic conjugation via uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1), conjugation into the bile via MRP2 represent the main action mechanism of clofibrate that turns it into the bilirubin conjugates and expels it from the bile. Bilirubin 60-69 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 214-220 34996412-1 2022 BACKGROUND: Pharmacogenetic (PGx) testing for germline variants in the DPYD and UGT1A1 genes can be used to guide fluoropyrimidine and irinotecan dosing, respectively. Irinotecan 135-145 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 80-86 35000550-3 2021 In vitro, CC-90001 glucuronidation was catalyzed by UGT1A9, UGT1A4, and UGT1A1, while oxidative metabolism was primarily mediated by CYP3A4/5 with minor contributions from CYP1A2, CYP2C9, CYP2B6, and CYP2D6.2. CC-90001 10-18 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 72-78 34718654-9 2022 Dasabuvir produced strong partial inhibition effect of UGT1A1 and UGT1A9 and relatively complete inhibition of UGT1A6. dasabuvir 0-9 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 55-61 35370268-0 2022 The Effect of Single-Walled Carbon Nanotubes on UDP-Glucuronosyltransferase 1A Activity in Human Liver. Carbon 28-34 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 48-78 35370268-4 2022 beta-Estradiol 3-glucuronidation, which is catalyzed mainly by UGT1A1, was inhibited by 99 and 76% in the presence of 0.1 mg/mL EC1.5-P- and FH-P-SWCNTs in human liver microsomes, respectively. Estradiol 0-14 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 35370268-4 2022 beta-Estradiol 3-glucuronidation, which is catalyzed mainly by UGT1A1, was inhibited by 99 and 76% in the presence of 0.1 mg/mL EC1.5-P- and FH-P-SWCNTs in human liver microsomes, respectively. 5-p 132-135 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 63-69 35370268-5 2022 The observed decrease of free UGT1A1 protein in the enzyme reaction mixture suggests a higher interaction with SWCNTs, and indicates the inhibition of beta-estradiol 3-glucuronidation. Estradiol 151-165 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 30-36 35356222-0 2022 UGT1A1 Allele Test Not Only Minimizes the Toxicity But Also Maximizes the Therapeutic Effect of Irinotecan in the Treatment of Colorectal Cancer: A Narrative Review. Irinotecan 96-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 0-6 35356222-2 2022 Adjusting the dose of irinotecan according to the uridine diphosphate glucuronosyltransferase (UGT) 1A1 genotype reflects the principle of individualized and precision medicine, and may improve the chemotherapy response and survival of CRC. Irinotecan 22-32 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 50-103 35356222-3 2022 Methods: To summarize the feasibility, efficacy and safety of high dose irinotecan in CRC patients with UGT1A1 wild-type or heterozygous alleles, PubMed, EMBASE, MEDLINE and the Cochrane Central Register of Controlled Trials online databases were searched from the date of creation to October 22, 2021. Irinotecan 72-82 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 104-110 35356222-5 2022 The results indicated that the maximum tolerated dose of irinotecan in CRC patients with UGT1A1 wild-type or heterozygous variant was significantly higher than the conventional recommended dose. Irinotecan 57-67 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 89-95 35356222-6 2022 Chemotherapy based on high dose irinotecan improved the clinical efficacy in mCRC patients with UGT1A1*28 wild-type and heterozygous variant, and the toxicity was tolerated, as reflected in most studies. Irinotecan 32-42 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 96-102 35356222-7 2022 Conclusions: We are optimistic about the application of high dose irinotecan for mCRC patients with UGT1A1*28 wild-type or heterozygous variant, which will provide a relatively clear direction for future research and certain norms for clinical practice. Irinotecan 66-76 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 100-106